targeting the heart using in vivo phage display

132
TARGETING THE HEART USING IN VIVO PHAGE DISPLAY by Maliha Zahid M.B., B.S., Aga Khan University, Pakistan, 1993 Submitted to the Graduate Faculty of Department of Human Genetics Graduate School of Public Health in partial fulfillment Of the requirements for the degree of Doctor of Philosophy University of Pittsburgh 2009

Upload: others

Post on 15-May-2022

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

by

Maliha Zahid

M.B., B.S., Aga Khan University, Pakistan, 1993

Submitted to the Graduate Faculty of

Department of Human Genetics

Graduate School of Public Health in partial fulfillment

Of the requirements for the degree of

Doctor of Philosophy

University of Pittsburgh

2009

Page 2: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

UNIVERSITY OF PITTSBURGH

Graduate School of Public Health

This dissertation was presented

by

Maliha Zahid

It was defended on

October 14th, 2009

and approved by

Dissertation Advisor: Paul D. Robbins, PhD

Professor Department of Microbiology and Molecular Genetics

School of Medicine University of Pittsburgh

Robert E. Ferrell, PhD

Professor Department of Human Genetics

Graduate School of Public Health University of Pittsburgh

Eleanor Feingold, PhD

Associate Professor Department of Human Genetics

Graduate School of Public Health University of Pittsburgh

Charles F. McTiernan, PhD

Associate Professor Department of Medicine

School of Medicine University of Pittsburgh

ii

Page 3: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

iii

Copyright © by Maliha Zahid

2009

Page 4: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

iv

Paul D. Robbins, PhD

TARGETING THE HEART USING IN VIVO PHAGE DISPLAY Maliha Zahid, PhD

University of Pittsburgh, 2009

Background: Ischemic heart disease remains the number one killer in the developed world. A

protein transduction peptide specific for the heart capable of efficiently delivering agents of

therapeutic potential at the time of injury, would be of immense public health significance. This

work was undertaken to identify peptide(s) able to transduce heart tissue in vivo in a tissue-

specific manner. Biopanning was performed in cell culture followed by in vivo with an M13

phage peptide display library. Using the heart-specific peptide, we delivered nemo-binding

domain peptide in a murine infarct model to test if NF-κB inhibition can reduce infarct size.

Methods and Results: A cardiomyoblast cell line, H9C2, was incubated with M13 twelve amino

acid phage peptide display library. Internalized phage was recovered, amplified and subjected to

a total of three rounds of in vivo biopanning where infectious, internalized phage was isolated

from cardiac tissue following intravenous injection. After the third round, 60% of sequenced

plaques carried the peptide sequence APWHLSSQYSRT, termed cardiac targeting peptide

(CTP). This peptide was synthesized either fluorescently labeled, biotinylated, or in combination

with a NEMO-binding peptide (NBD), an inhibitor of the inducible NF-kappa B Kinase (IKK).

We demonstrate that CTP was able to transduce cardiomyocytes functionally in culture in a

concentration and cell-type dependent manner. Mice injected with CTP showed significant

transduction of heart tissue with minimal uptake by lung and kidney capillaries, and no uptake in

Page 5: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

v

liver, skeletal muscle, spleen or brain. The level of heart transduction by CTP also was not

observed with a cationic transduction domain. CTP-NBD was able to inhibit NF-κB activation in

cell culture in a dose-dependent fashion. When administered to mice in a murine infarct model,

CTP-NBD showed a trend towards smaller infarct size, which did not reach statistical

significance.

Conclusions: Biopanning using a peptide phage display library identified a peptide, termed CTP,

able to transduce cardiomyoblast cell line in vitro, and heart tissue in vivo, efficiently and

specifically. Administration of CTP-NBD to post-infarct mice showed a trend towards reduction

of infarct size. CTP could be used to deliver therapeutic peptides, proteins and nucleic acid

specifically to the heart.

Page 6: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

vi

TABLE OF CONTENTS

PREFACE ........................................................................................................................................ XII

LIST OF ABBREVIATIONS ............................................................................................ XIV

1.0 INTRODUCTION ............................................................................................................. 1

1.1 SCOPE OF THE PROBLEM ................................................................................. 1

1.2 PROTEIN TRANSDUCTION DOMAINS .......................................................... 2

1.3 BIOPANNING OR PHAGE DISPLAY ................................................................ 3

1.4 NUCLEAR FACTOR ΚB ..................................................................................... 10

1.4.1 Classical NF-κB Activation Pathway .............................................................. 12

1.4.2 Alternate NF-κB Activation Pathway ............................................................. 15

1.5 NEMO-BINDING DOMAIN PEPTIDE ............................................................. 18

1.6 NF-ΚB AND THE HEART................................................................................... 21

2.0 PHAGE DISPLAY TO TARGET THE HEART....................................................... 25

2.1 PHAGE DISPLAY ................................................................................................. 25

2.2 IN VITRO TRANSDUCTION EXPERIMENTS: CONFOCAL STUDIES . 33

2.3 IN VITRO TRANSDUCTION: FUNCTIONAL ASSAY ................................. 39

2.4 IN VIVO TRANSDUCTION STUDIES .............................................................. 45

2.5 HUMAN HEART TISSUE EXPERIMENTS .................................................... 57

2.6 CHAPTER SUMMARY ........................................................................................ 61

Page 7: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

vii

3.0 MURINE INFARCT SIZE STUDIES USING NBD ................................................. 64

3.1 GENERATION OF THE ANIMAL MODEL OF MYOCARDIAL

INFARCTION .......................................................................................................................... 65

3.2 INFARCT STUDIES USING 8K-NBD ............................................................... 67

3.3 INFARCT STUDIES USING CTP-NBD ............................................................ 75

4.0 SUMMARY, IMPLICATION OF OUR FINDINGS AND FUTURE

DIRECTIONS ................................................................................................................................... 83

4.1 SUMMARY OF OUR FINDINGS....................................................................... 83

4.2 IMPLICATIONS AND FUTURE DIRECTIONS ............................................ 85

5.0 METHODS ....................................................................................................................... 91

5.1 PROTOCOL FOR PEPTIDE PHAGE DISPLAY ........................................... 91

5.1.1 Materials .............................................................................................................. 92

5.1.2 Methods................................................................................................................ 95

5.2 IN VITRO TRANSDUCTION EXPERIMENTS............................................ 103

5.2.1 Cell transduction experiments with CTP-6CF ............................................ 103

5.2.2 8% Paraformaldehyde stock solution ........................................................... 104

5.3 METHODOLOGY FOR TRANSFECTION OF CELLS AND INHIBITION

OF NF-ΚB ACTIVATION ................................................................................................... 105

5.3.1 Materials ............................................................................................................ 105

5.3.2 Methods.............................................................................................................. 106

5.4 MOUSE INFARCT MODEL ............................................................................. 107

BIBLIOGRAPHY ........................................................................................................................... 110

Page 8: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

viii

LIST OF TABLES

Table 1. Summary of studies utilizing NBD peptide. ...................................................................... 19

Table 2. Summary of NF-kappa B studies with cardiac ischemia-reperfusion injury or

myocardial infarction. ......................................................................................................................... 23

Table 3. Number of phage injected and recovered from each cycle of phage display ................... 29

Page 9: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

ix

LIST OF FIGURES

Figure 1. Schema of available display technologies. ......................................................................... 4

Figure 2. Structure of M13 bacteriophage. ......................................................................................... 7

Figure 3. Assembly of phage virion and its release from E. coli cells. ............................................. 7

Figure 4. Schematic representation of steps in screening a peptide phage display library. ............. 9

Figure 5. Activation of classical NF-κB signaling by TNF-a. ......................................................... 14

Figure 6. Activation of NF-κB signaling by CD40. ......................................................................... 16

Figure 7. Experimental design of the combinatorial in vitro and in vivo phage display. ............... 28

Figure 8. Recovered phage normalized to tissue weight and plotted as a ratio of heart to kidney

(a) and output to input ratios (b). ....................................................................................................... 30

Figure 9. Physical properties of Cardiac Targeting Peptide or CTP. .............................................. 33

Figure 10. Confocal micrographs of H9C2, 3T3, MCA205, HeLa and HK-2 cells incubated with

increasing concentrations of CTP-6CF; Nuclei - blue (DRAQ5), CTP-6CF - green, 20x. Scale

bars represent 100uM. ........................................................................................................................ 36

Figure 11. High magnification confocal micrographs of H9C2 cells incubated with CTP-6CF for

30 (top row) and 60 minutes (bottom row). ...................................................................................... 37

Figure 12. Live cell imaging of H9C2 cells incubated with RAN-6CF, CTP-6CF or TAT-6CF. 38

Figure 13. Experimental protocol for the in vitro transduction experiments using Luciferase

readout. ................................................................................................................................................ 40

Page 10: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

x

Figure 14. Inhibition of Luciferase activity by CTP-NBD in H9C2 cells. ..................................... 41

Figure 15. Lack of inhibition of Luciferase activity by CTP-NBD in MCA205 cells. .................. 42

Figure 16. Relative lack of inhibition of Luciferase activity by CTP-NBD in HeLa cells. ........... 43

Figure 17. H9C2 cell viability after transfection, treatment with CTP-NBD and subsequent TNF-

alpha treatment. ................................................................................................................................... 44

Figure 18. Transduction of mouse heart by intravenously injected CTP-6CF at 15 minutes. ....... 46

Figure 19. Quantification of fluorescence in mouse hearts (n=3 in each group). .......................... 47

Figure 20. Transduction of heart tissue after intravenous injection with CTP-biotin-streptavidin-

Alexa488 complex. ............................................................................................................................. 50

Figure 21. Biodistribution of CTP-biotin-streptavidin-Alexa488 complex after intravenous

injection ............................................................................................................................................... 50

Figure 22. In vivo, whole mouse imaging after intra-cardiac injection of CTP, RAN or PBS

conjugated to fluorescent 40nm beads. ............................................................................................. 52

Figure 23. Confocal micrographs of hearts injected with PBS+fluospheres, RAN+fluospheres or

CTP+fluospheres. ............................................................................................................................... 53

Figure 24. Uptake of CTP-6CF preferentially by normal myocardium with exclusion from the

infarct area. .......................................................................................................................................... 55

Figure 25. Comparison of heart tissue transduction between CTP, 8K and RAN peptide. ........... 56

Figure 26. Transduction of human heart tissue ex vivo by CTP-6CF. ............................................ 58

Figure 27. Results of ex vivo human heart tissue experiment at 30 mins. ...................................... 60

Figure 28. Inhibition of LPS induced Luciferase signal by 8K-NBD ............................................. 69

Figure 29: Post-operative mortality by treatment group in the 8K-NBD study. ............................ 70

Page 11: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

xi

Figure 30. Heart weight to body weight ratios across the treatment groups in the 8K-NBD study.

.............................................................................................................................................................. 71

Figure 31. Infarct size across different treatment groups in the 8K-NBD study. ........................... 72

Figure 32. Representative confocal micrographs of TUNEL staining in each treatment group in

the 8K-NBD study. ............................................................................................................................. 73

Figure 33. Quantification of TUNEL positive data in each treatment group in 8K-NBD study. .. 74

Figure 34. Post-operative mortality by treatment group in the CTP-NBD study. .......................... 76

Figure 35. Infarct size by treatment groups in the CTP-NBD study. .............................................. 77

Figure 36. Transduction of the mouse heart by the D-CTP-NBD-SA488 complex. ..................... 79

Figure 37. Inhibition of NF-κB activation by the D-isoform of CTP-NBD. .................................. 80

Figure 38. Infarct sizes in the two treatment groups. ....................................................................... 81

Figure 39. Temperature dependency of CTP's transduction ability. ............................................... 86

Figure 40. H9C2 cell viability after incubating with increasing concentrations of CTP-NBD. .... 87

Figure 41. H9C2 cells transfected with different AAV viral vectors expressing EGFP. ............... 90

Page 12: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

xii

PREFACE

Lay Summary: Heart disease remains the number one killer in the US as well as the rest of the

developed world, and is increasing steadily in the developing world with its industrialization.

Hearts attacks are the commonest manifestation of it and occur when an atherosclerotic plaque

ruptures, exposing a rough surface to the blood leading to blood clotting and the resulting clot

completely occluding the said artery. This leads to distal heart muscle tissue being deprived of

oxygen and injury, which if it continues unchecked, leads to irreversible muscle death. Current

therapies revolve around opening the clogged artery as expeditiously as possible, either with clot

busting drugs or catheters. Opening the artery exposes the jeopardized muscle to a surge of

oxygen and generation of molecules which are harmful to the muscle as well. Many molecules

and therapies have been tested in animal models that have shown efficacy in preventing this, but

all suffer from the drawback of lack of an appropriate delivery model. The pathology outlined

above occurs within hours of the ruptured plaque and the window of opportunity is measured in

hours, with no benefit seen past 12 hours. Therefore a delivery vehicle would have to target the

heart efficiently, within hours, to deliver ready-to-go peptides or proteins of therapeutic

potential. Also such a mode of delivery should be specific to heart in order to reduce the amount

of peptide/protein needed to see a beneficial effect and to potentially decrease side effects. In the

work I will be presenting in this thesis, we have done just that. Using the well-established

technique of phage display, we have identified a peptide that targets the heart efficiently,

Page 13: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

xiii

specifically, and very rapidly, within 30 minutes. This peptide can be used as a carrier to deliver

peptides or full-length proteins to achieve therapeutic benefit in reducing heart attack sizes and

perhaps even for diagnostic purposes.

Page 14: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

xiv

LIST OF ABBREVIATIONS

6CF 6-carboxyfluoroscein

8K Homopolymer of 8 Lysine Residues

6R Homopolymer of 6 Arginine Residues

CPP Cell Penetrating Peptide

CTP Cardiac Targeting Peptide

Da Daltons

EBD Evans Blue Dye

EGFP Enhanced Green Fluorescent Protein

IKK I-KappaB Kinase

IκBα I-kappa B-alpha

IκBβ I-kappa B-beta

IκBγ I-kappa B-gamma

IL-1 Interleukin 1

IM Intramuscular

IP Intraperitoneal

IR Ischemia-reperfusion

KO Knock-out

LPS Lipopolysaccharide

MI Myocardial Infarction

Mut Mutant

NBD Nemo Binding Domain

NEMO NF-kappa B Essential Modulator

Page 15: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

xv

NF-κB Nuclear Factor-kappa B

PBS Phosphate Buffered Saline

PFA Paraformaldehyde

PTDs Protein Transduction Domains

RAN Random Peptide

RT Room Temperature

SA-488 Streptavidin-Alexa 488

SD Standard Deviation

SEM Standard Error of the Mean

TAT Transactivator of Transcription

TNFα Tumor Necrosis Factor-alpha

Page 16: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

1

1.0 INTRODUCTION

1.1 SCOPE OF THE PROBLEM

Ischemic heart disease and occlusive coronary artery disease continue to be the number one killer

in the developed world. There are an estimated 500,000 acute ST-elevation myocardial

infarctions (MI) in the US alone each year [1], and this is becoming an increasingly significant

problem in the developing world [2]. Current approaches for management of an acutely occluded

coronary artery leading to an MI consist of anti-platelet and anti-thrombotic strategies with

intervention aimed at opening the infarct-related artery in a timely fashion. Although this

approach is able to protect cardiomyocytes from necrosis, with resulting decrease in morbidity

and mortality, it necessitates exposing the heart to post-ischemic reperfusion injury. Limiting this

reperfusion injury and decreasing apoptosis would ultimately lead to greater myocardial salvage

and prevention of development of heart failure.

Numerous animal studies have identified biological agents able to ameliorate this

ischemia-reperfusion injury and reduce the ultimate infarct size [3-5]. The clinical application of

these potentially effective biological therapies for cardiac conditions, like myocardial infarction,

has been limited by efficiency and specificity of delivery of therapeutic agents. For example, for

gene therapy approaches, plasmid delivery to the heart is very inefficient whereas there are

significant time delays associated with cardiac gene delivery using viral-based vectors in

Page 17: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

2

addition to issues of pre-existing neutralizing antibodies or exciting an immune responses to

certain viral vectors. The well-characterized cell penetrating peptides, like TAT from HIV coat

protein, homopolymers of arginine or lysine, are not cell specific and transduce hepatocytes and

multiple other organs in addition to the heart. Identifying a peptide with transduction capabilities

specific for the heart would allow for new approaches for effective cardiac delivery of

therapeutics to be developed.

1.2 PROTEIN TRANSDUCTION DOMAINS

Protein transduction domains (PTD) or cell penetrating peptides (CPP) are small peptides able to

transverse plasma membranes and carry peptides, full-length proteins, oligonucleotides, iron

nanoparticles and liposomes as “cargoes”. These PTDs can be broadly classified under three

classes; cationic or positively charged PTDs, hydrophobic or protein leader sequence-derived

domains and peptides identified by phage display that are able to transduce cells in a cell-type

specific manner. Interest in these moieties began with the simultaneous reports by Franckel and

Pabo [6], and Green and Lowenstein [7], that HIV-1 TAT (transactivator of transcription) protein

was able to cross plasma membranes. A few years following these initial reports, the

Antennapedia homeodomain of Drosophila melanogaster was shown to act in a similar manner

[8]. Further investigation revealed that smaller peptides derived from these larger full-length

proteins were responsible for their transduction abilities and could carry multiple, different, full-

length proteins as cargoes [9-11]. Interest in these unique peptides has been burgeoning leading

to identification of multiple naturally occurring [12-14] as well as engineered PTDs [15-16].

Page 18: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

3

Multiple studies have shown the ability of PTDs to deliver peptides of therapeutic

potential [17-18], full-length proteins [11, 19], oligonucleotide [20], 40nm iron nanoparticles

[21-22], drugs [23], and liposomes [24]. In addition there is intense interest in developing PTDs

as vehicles for delivery of small interfering RNA (siRNA) in vitro [25-26] and in vivo [27-28],

with novel strategies aimed at PTD and siRNA complex escaping the endosomal compartment

[29-30] or increasing efficacy by avoiding the nuclear compartmentalization [30]. These selected

number of publications, involving myriad applications of PTDs, highlight not only the interest

but also the potential application of PTDs as delivery tools in a variety of disease models

employing many different strategies of therapeutic potential. Although PTDs have shown the

ability to deliver biologically active cargo in vivo, and even cross the blood-brain barrier [11],

the lack of cell or tissue specificity seen with the cationic or hydrophobic PTDs, limits their

therapeutic potential and would add to their toxicity limiting use in vivo. In light of these

considerations, it has become increasingly apparent that for PTDs to be useful, it is necessary to

identify peptides that are tissue specific in order to limit toxicity and enhance therapeutic

potential. To identify such peptides, biopanning or phage display has emerged as a powerful

technique.

1.3 BIOPANNING OR PHAGE DISPLAY

The principal underlying display technologies consist of the ability to physically link phenotypes

of polypeptides displayed on a certain platform to their corresponding genotype. Typically the

phenotype registered in display experiments is through physical binding and serial selection.

Figure 1 shows the schema of available display technologies [31].

Page 19: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

4

CLOSE

Figure 1. Schema of available display technologies.

Page 20: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

5

All display platforms are based on the ability to physically link the polypeptide produced

by a library clone to its corresponding genotype. This allows one to recover the DNA encoding

the clone selected based on the desired polypeptide phenotype, such as binding to the target.

The ability to modify filamentous bacteriophage to express polypeptides on the surface as

part of the bacteriophage’s coat protein was first reported by Smith [32] in 1985. This technique

was originally developed to map epitope-binding sites of antibodies by mapping against

immobilized immunoglobulins. This seminal work led to phage display being utilized as a

powerful method to detect polypeptide binding with a diverse range of biological, as well as

technical and medical applications [33].

A brief word on the biology of phage is warranted here. “Phages” or bacteriophage are

prokaryotic bacterial viruses, which are the most abundant biological entities in the environment

with estimates ranging from 1030 to 1032 and play a key role in many biological systems.

Bacteriophages as a group are extremely diversified. The number of known phage has been

expanding for decades at a rate of ~100/year. The Felix d’Herelle Reference Center for Bacterial

Viruses is charged with keeping track of these. The first survey was published in 1967 with latest

surveys made in 1995 and 2000 [34-35]. Phages are broadly classified as Tailed, Polyhedral,

Filamentous or Pleomorphic. According to the latest electron microscopic analysis of over 500

phages [36], the vast majority (~96%) are tailed with polyhedral, filamentous and pleomorphic

phage comprising 3.7% of the total number of phage studied. Of these, filamentous phage has

been studied widely for the purpose of phage display. Filamentous phage are non-lytic and leave

the host by budding through the host plasma membrane without killing their pilli-bearing host

bacteria, though markedly slowing it’s rate of replication. The filamentous phage best studied

Page 21: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

6

biochemically and genetically is the M13 and its close relative fd bacteriophage. Both are F-

specific phages that infect Escherichia coli bacteria.

M13 phage is a long, thread shaped particle, 6.5nm in diameter and 900nm in length. Its

genome is a single-stranded, circular DNA containing 6,407 nucleotides protected by a long

cylindrical protein coat. The most abundant coat protein is the major coat protein pVIII, of which

~2700 molecules are present on the coat. There are only 5 copies of the minor coat protein pIII at

one end of the phage particle, which along with 5 molecules of another minor coat protein, pVI,

is involved in bacterial cell binding. At the other end of the phage are minor coat proteins pVII

and PIX, which are needed for initiation and maintenance of phage assembly inside the host.

During the assembly process the resulting fusion coat proteins are transported to the bacterial

periplasm or inner cell membrane and incorporated into a new phage particle along with the

single stranded DNA genome of the phage carrying the genotypic information for the displayed

fusion protein. Figure 2 is a schematic representation of phage structure, followed by a

diagrammatic representation of phage assembly and budding from an E. coli host cell.

Page 22: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

7

• Dimensions:– 6.5nm in diameter– Length dependant on genome

but wild type approximately 930nm

– Mass 16.3MD of which 87% comprised of protein

• Genome:– Single stranded circular DNA

molecule, covalently closed– Housed in a flexible protein

cylinder

Figure 2. Structure of M13 bacteriophage.

Figure 3. Assembly of phage virion and its release from E. coli cells.

Page 23: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

8

Libraries have been constructed utilizing virtually all of the coat proteins of filamentous

phage modified for the purpose of phage display. However the commonest filamentous phage

libraries consist of phage with modified minor capsid protein pIII, expressing 109 peptides, or

more, of different permutations. Filamentous phage infect pilus-positive bacteria that are not

lysed by the infecting phage and secrete multiple, identical copies of it displaying a particular

insert. Phage display constitutes exposing the target of interest to a large, randomized library of

phage expressing peptides on a surface coat protein. After binding, the non-relevant phage is

washed away, the bound phage eluted, expanded in pilli-positive host bacteria, and re-exposed to

the target of interest. Four to six cycles of biopanning lead to adequate enrichment with the

relevant clone phage, which can subsequently be sequenced, and the peptide motif carried as a

fusion coat protein identified. This technique allows one to identify peptides able to bind

immobilized targets, surfaces of different cell types and even target tissues in vivo without a

priori knowledge of the target ligand, as outlined in the figure below.

Page 24: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

9

Figure 4. Schematic representation of steps in screening a peptide phage display library.

Initial studies utilizing phage display libraries were limited to in vitro work, phage

display being carried out against immobilized antigen targets or cell-specific ligands. Arap and

colleagues [37] showed the feasibility of in vivo phage display leading to identification of

peptides homing to tumor vasculature as well as the ability to use these peptides to target anti-

Page 25: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

10

cancer therapy. Further in vivo work has clarified that not only tumor vasculature but also normal

organs, like adipose tissue [38] and the heart [39], among other organs [40-41], bear ligands that

can be targeted by in vivo phage display. Work in our lab has identified peptides targeting

synoviocytes which are able to deliver therapies to synovial fibroblasts in vivo [42] as well as

protect islet cells in a mouse model of diabetes [43]. Hence biopanning or phage display has

evolved as a powerful technique for identification of peptides targeting not only immobilized

antigens, but cell-specific ligands in vitro, vasculature of various organs and tissue types in vivo.

1.4 NUCLEAR FACTOR ΚB

NF-κB was first identified as a transcription factor binding to the intronic enhancer of the Кappa

light chain gene in B-cells over 20 years ago [44-45]. It soon became apparent that this

transcription factor plays a major role in orchestrating innate and adaptive immune responses

that are key to a multicellular organism’s ability to respond to environmental, mechanical,

chemical and microbial stresses. It also became apparent that deregulation of this transcriptional

activity can lead to a myriad of seemingly different autoimmune diseases like rheumatoid

arthritis, inflammatory bowel diseases, multiple sclerosis as well as cancer. A Pub Med search

today would reveal over 30,000 papers. Therefore the review that follows will, out of necessity,

be kept brief, and to the point as it relates to the thesis work done.

NF-κB transcription factor is a family of five cytosolic proteins; p50, p52, p65 (RelA), c-

Rel, RelB, encoded by NFKB1, NFKB2, RELA, REL, and RELB respectively. All of them share

an N-terminal Rel-homology domain (RHD) that is responsible for DNA binding and homo- or

hetero-dimerization. RelA, c-Rel and RelB also contain C-terminal transcription activation

Page 26: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

11

domains (TAD), whereas p50 and p52 do not and depend on heterodimerization with other

subunits to lead to activation of transcription. NF-κB dimers bind to specific κB consensus

sequences (5` - GGGRNWYYCC – 3`; where N=any base, R=purine, W=adenine or thymine,

and Y=pyrimidine) within the promoter-enhancer regions of target genes and regulate

transcription through recruitment of co-activators and repressors. These proteins reside in the

cytosol in an inactive state either bound to inhibitors called IκBs or as the unprocessed or

precursor proteins p100 and p105. In response to stimuli, p100 and p105 are processed to form

p52 and p50 respectively. Three “classical” proteins, IκBα, IκBβ and IκBε, are characterized by

the presence of multiple ankyrin repeats that mediate binding of NF-κB dimers and interfere with

the nuclear localizing signal of the latter, thus keeping these proteins cytosolic and hence

inactive.

In response to appropriate stimuli, NF-κB activation occurs, leading to its nuclear

accumulation. Activation of NF-κB by phosphorylation and degradation of inhibitory IκBs

occurs in the classical or canonical pathway that comes into play in response to TNF-α or IL-1

binding to their receptors. The classical pathway involves phosphorylation of the IκB proteins at

specific serine residues (phosphorylation of IκBα on serine residues 32 and 36, and

phosphorylation of IκBβ on serine residues 19 and 23), marking the protein for ubiquitination

and subsequent degradation, with release of the inhibitory influence on the NF-κB dimers and

their subsequent activation. This pathway centers on activation of the trimeric IκB kinase

complex, (IKK), comprising the catalytic subunits IKKα, IKKβ and the non-catalytic subunit,

IKKγ (also called NEMO for NF-κB essential modulator). IKKα and IKKβ share 50% sequence

identity with 65% sequence identity of their catalytic subunits. There are some notable

differences with IKKα having a nuclear localizing signal that is unique to it and IKKβ having a

Page 27: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

12

C-terminal NEMO-binding domain that exhibits much greater affinity for IKKγ/NEMO than

IKKα. IKKγ or NEMO is obligatory for classical NF-κB activation as in its absence IKK

complex can no longer be formed or activated [46-47]. Of the two catalytic subunits, IKKβ is the

major IκB kinase in most cell types with few exceptions, and plays a central role in the canonical

activation pathway. In contrast IKKα is essential for activation of the alternate pathway.

1.4.1 Classical NF-κB Activation Pathway

As mentioned above, NF-κB activation can occur via the canonical or classical pathway, or the

alternate pathway. Activation via the canonical pathway requires activation of the IKK complex.

The three major IKK proteins consist of IKKα, IKKβ and IKKγ (or NEMO- NF-κB essential

modulator) which dimerize through the leucine zipper domain which is also required for kinase

activity. Both IKKα and IKKβ bind the third protein component of the IKK complex, NEMO,

through the C-terminal hexa-peptide, NEMO-binding domain (NBD; Leu-Asp-Trp-Ser-Trp-

Leu). Competition experiments and biophysical analyses using the NBD peptide indicate that

IKKβ binds to NEMO with higher affinity than IKKα. Activation of the IKK complex requires

phosphorylation of at least one of the IKK subunits. Active IKKβ is phosphorylated on two

serines, Ser177 and Ser181 within the activation loop of the kinase domain. Similarly IKKα is

phosphorylated on activation loop serine residues 176 and 180. The exact mechanism of this

activation, autophosphorylation or an upstream IKK kinase mediated phosphorylation, remains

to be elucidated.

Much of the current understanding of the classical pathway of NF-κB activation comes

from studies of TNF-α signaling. TNF-α binds to two receptors, TNFR1 and TNFR2, of which

TNFR1 binding plays a much broader role in NF-κB activation. Binding of TNF-α to its receptor

Page 28: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

13

TNFR1 leads to recruitment of TNFR1-associated death domain protein (TRADD). Receptor

engagement also results in a biphasic ubiquitination of RIP1, a member of related protein

kinases. However studies of RIP1 deficient cells show that it is likely needed for a docking

action for IKK through its interaction with IKKγ [48]. How this leads to IKK activation is

unclear but one mechanism proposed is recruitment of MEKK3, a kinase to the TNFR1 signaling

complex, leading to phosphorylation of the IKKα and IKKβ activation loops [49]. However

MEKK3 is not unequivocally an essential IKK kinase and there are likely many others, like

TAK1, and the relative importance/role of an IKK kinase is likely to be cell-dependent. TAK1

might be recruited to the TNFR1 signaling complex via TRAF-2 dependent polyubiquitination of

RIP1 or by more direct recruitment of TAK1 via TRAF2 or TRAF6. Although the exact nature

and events leading to IKK activation differ depending on the stimulus (TNF-α , IL-1, antigen

binding etc), the ultimate common path is activation of the IKKα and/or IKKβ by

phosphorylation of serine residues in the activation loop. This activation of the IKK complex

leads to phosphorylation of IκBα, leading to its proteasomal degradation and release of its

inhibition of p50/p65 heterodimers, their nuclear translocation and initiation of transcriptional

activity. This is schematically represented in the figure below (Figure 5) for TNF-α mediated

NF-κB activation [50].

Page 29: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

14

Figure 5. Activation of classical NF-κB signaling by TNF-a.

Page 30: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

15

Ligation of TNFR1 results in TRADD-dependent TRAF2/TRAF5 and RIP1 recruitment.

TRAF2 causes K63-linked ubiquitination of RIP1 and also recruits IKK to the receptor complex,

where binding of IKKγ to ubiquitinated RIP1 stabilizes IKK interaction with the receptor

complex. This may also lead to a conformational change of the IKK complex. TAB2 and TAB3

interact with TRAF2 and TAK1, leading to TAK1 activation that may then phosphorylate IKKβ.

Alternatively, MEKK3, which is brought near the receptor complex presumably by RIP1 may

also phosphorylate and activate IKK. On the other hand, IKK may also be activated by

autophosphorylation. Activated IKK phosphorylates IκBα at specific serine residues, leading to

the proteasome-mediated degradation of the latter. Degradation of IκBα releases the NF-κB

heterodimers, which then migrate to the nucleus and regulate gene expression.

1.4.2 Alternate NF-κB Activation Pathway

NIK is an unstable protein, subject to rapid turnover in non-stimulated cells, first identified as a

protein kinase, whose over-expression results in NF-κB activation [51]. Stimulation of B cells

with certain TNF family members, such as CD40L, results in stabilization of NIK trans-

autophosphorylation, its subsequent accumulation leading to activation of IKKα. In the resting

state, NIK is associated with TRAF2, TRAF3 and cIAP1/2. TRAF3 appears to function as an

adapter linking NIK to the ubiquitin ligase complex consisting of TRAF2/cIAP1/1. This complex

causes ubiquitination of NIK and consequent degradation. Upon cell stimulation with appropriate

receptor-ligand binding, a protein ubiquitination cascade ensues with TRAF2 ubiquitinates

cIAP1/2 through a K63-linkage leading to the activation of their K48-specific ubiquitin ligase

activity towards TRAF3. This cascade leads to NIK stabilization, accumulation, and activation of

IKKα through phosphorylation, leading to processing of p100 to its p52 residue and release of

Page 31: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

16

RelB/p52 heterodimers allowing for nuclear translocation, as demonstrated in Figure 6 below

[50].

Figure 6. Activation of NF-κB signaling by CD40.

In resting cells (left), p100, via its C-terminal ankyrin repeats, binds and keeps RelB in

the cytosol. NIK, which is an essential kinase involved in the phosphorylation and proteasome-

Page 32: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

17

mediated C-terminal processing of p100, is maintained at very low levels owing to rapid

proteasome-dependent degradation. TRAF3 links NIK to an E3 complex containing TRAF2 and

cIAP1/2, thereby promoting cIAP1/2-mediated K48-linked NIK polyubiquitination and

proteasomal degradation. TRAF3 also undergoes a low level of K48-linked polyubiquitination

under resting conditions. Activation of CD40 by CD40L (right) leads to recruitment of the

cIAP1/2:TRAF2:TRAF3 complex to the receptor, where cIAP1/2 undergoes TRAF2-dependent

K63-linked polyubiquitination. TRAF2 also undergoes K63-linked self-ubiquitination. K63-

linked ubiquitination of cIAP1/2 enhances their K48-specific E3 ubiquitin ligase activity toward

TRAF3, leading to proteasomal degradation of the latter. As a result, TRAF3 levels in the cell

drop below a critical threshold, and NIK can no longer be recruited to the cIAP1/2:TRAF2

complex. This leads to stabilization and accumulation of newly synthesized NIK and its

activation presumably via autophosphorylation, resulting in activation of IKKα. Activated IKKα

phosphorylates p100, leading to proteasome-mediated processing of p100 to p52. C-terminal

truncation of p100 releases the p52:RelB heterodimer, which migrates to the nucleus and

regulates transcription of its target genes.

Once activated, the termination of active NF-κB heterodimers is poorly understood.

Studies have focused on upstream targets and shown that among the myriad of genes activated

by NF-κB, IκB protein synthesis is up regulated, thus serving as a negative feedback loop.

However terminating the activity of DNA-promoter bound NF-κB remains poorly understood

and is a subject of active ongoing research.

Page 33: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

18

1.5 NEMO-BINDING DOMAIN PEPTIDE

As detailed above, activation of NF-κB, via the canonical pathway, requires activation of the

IκB-Kinase, or IKK complex. This complex consists of three major proteins, IKKα, IKKβ and

IKKγ, the latter also known as NEMO (NF-κB essential modulator). An elegant study by May et.

Al. [52] revealed that this interaction occurs not only between IKKβ and NEMO, but also IKKα

and NEMO, albeit with lesser affinity. This study also mapped the region interacting with

NEMO to the NH2-terminal α-helical region of both IKKα and IKKβ, consisting of 6 amino

acids, Leu-Asp-Trp-Ser-Trp-Leu, and was termed the nemo-binding domain (NBD). In the study

a cell permeable peptide was created consisting of the Antennapedia homeodomain, a PTD,

fused to the 11 amino acids from T735 to E745 of IKKβ. This peptide was termed NBD peptide.

A mutant NBD peptide was also generated with W739 and W741 replaced with alanines. The

NBD peptide inhibited in vitro interaction of IKKβ with NEMO and disrupted formation of

endogenous IKK complexes in HeLa cells. Pre-treating HeLa cells with this NBD peptide

inhibited TNF-α induced NF-κB activation. Interestingly, the basal NF-κB activity was enhanced

approximately two-fold by the NBD peptide, suggesting that removal of NEMO slightly

increases the basal activity of the IKK complex. The differential binding abilities of IKKα and

IKKβ with NEMO were further characterized by mutational analysis showing that for IKKβ

nemo-binding domain to interact with NEMO, the aspartate residues at 738 and two tryptophan

residues at 739 and 741 were critical [53]. The requirement for binding to IKKα was not equally

stringent, with the first tryptophan residue at position 740 being most important.

Since the description of the NEMO-binding domain and generation of a small peptide

able to inhibit IKK complex activation, this peptide has been studied in numerous animal models

of inflammatory diseases. The original report detailed efficacy of this peptide in reducing

Page 34: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

19

phorbol 12-myristate 13-acetate induced ear edema, repressing E-selectin expression by HUVEC

cells in response to TNF-α, as well as decreasing peritonitis in mice injected with intraperitoneal

zymosan [52]. Following these initial reports, the NBD peptide in association with various

protein transduction domains (PTD-5, TAT or Antennapedia homeodomain), has been shown to

protect pancreatic islet cells from detrimental effects of IL-1β [43], block osteoclastogenesis,

synovial inflammation and bone erosion in inflammatory arthritis [54-55], and be beneficial in

two different models of murine colitis [18, 56]. In addition to murine models of inflammatory

bowel disease, NBD peptide was able to decreased bowel injury and decrease mortality in a

neonatal rat model of necrotizing enterocolitis [57]. The table below summarizes the studies that

have been undertaken to date utilizing NBD peptide.

Table 1. Summary of studies utilizing NBD peptide.

Authors Journal/Year Model Result

Long, YM et. Al.

[58]

W J Gastro/2009 Rat/Pancreatitis ↓ Inflammation

Ianaro, A et. Al.

[59]

Canc Lett/2009 Melanoma cell line ↓ Proliferation

Dave, SH et. Al.

[18]

J Immun/2007 Mice/Colitis ↓ Inflammation/colitis

Tapia, MA et. Al.

[60]

Cell Cycle/2007 Breast cancer cell line ↑Doxorubicin

sensitivity

Shibata, W et. Al.

[56]

J Immun/2007 Mice/Colitis ↓ Inflammation/colitis

De Plaen, IG et. Pediat Res/2007 Rat/Necrotizing colitis ↓ Bowel Injury

Page 35: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

20

Al. [57]

Tas, SW et. Al.

[61]

Arthir Res Ther/2006 Rat/Arthritis ↓ Arthritis Severity

van den Tweel,

ER et. Al. [62]

Pediat Res/2006 Rat/Cerebral ischemia ↑ Neuronal Damage

Di Meglio, P et.

Al. [55]

Arthrit rheum/2005 Mice/Carrageenan-

induced paw edema

↓ Paw Edema

Dai, S et. Al. [54] JBC/2004 Mice/Inflammatory

arthritis

↓Inflammation

↓erosions

Biswas, DK et.

Al. [63]

PNAS/2004 Breast cancer cell line ↑Apoptosis/↓cell

proliferation

Ashikawa, K et.

Al. [64]

Bioch Pharma/2004 Lymphoid/myeloid

cell line

↓Doxorubicin

sensitivity

Rehaman, KK et.

Al. [43]

JBC/2003 Mouse islet cell line ↑Viability

↑islet cell function

Thomas, RP et.

Al. [65]

Surgery/2002 Pancreatic cancer cell

line

↑Apoptosis/↓cell

viability

From the foregoing studies, it would appear that the NBD peptide, with an appropriate

protein transduction domain acting as a delivery vehicle, could be of use as a beneficial, anti-

inflammatory agent.

Table 1 - continued

Page 36: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

21

1.6 NF-ΚB AND THE HEART

Many of the key elements of the canonical NF-κB pathway have been identified in cardiac

myocytes. Exposing cardiac cells to stimuli like LPS, cytokines or reactive oxygen species as

well as ischemia-reperfusion leads to phosphorylation and subsequent degradation of IκBα. The

cytoplasmic to nuclear shuttling of p65/p50 heterodimers has also been confirmed in cardiac

myocytes, using electrophoretic mobility shift assays that employed κB sequence-containing

probes that confirmed the identity of the proteins within these complexes [66-68]. Of the 5 NF-

κB family proteins, only p65, p50, p52 and RelB have been identified in cardiac myocytes thus

far. These family members can form heterodimers and act as transcription factors, or p50 can

homodimerize to act as a repressor of transcription. Although detailed studies of the dimeric

species present in the heart have not been performed, it would appear that NF-κB-dependent

transcription activation or repression is regulated primarily by p65/p50 or p50/p50 dimers

respectively [69-72]. As detailed earlier, NF-κB dimmers reside in the cytoplasmic compartment

and are kept inactive by their association with inhibitor IκB proteins. Of the 8 IκB family

members, only IκBα, IκBβ, IκBε, Bcl-3, p100 and p105 have been identified in the heart.

Studies have revealed a role for NF-κB stimulation in macrophages in atherosclerotic

plaques [73-75] as well as inhibition of endothelial NF-κB protecting mice from atherosclerosis

[76]. In addition several studies have high-lighted the importance of NF-κB in generation of

cardiac hypertrophy in response to mechanical stretch [77-78], transverse aortic banding [79], in

response to Angiotensin II [80] with blockade of NF-κB in transgenic IκBα dominant-negative

mice or RNA interference leading to prevention of cardiac hypertrophy and heart failure [81-84].

The role of NF-κB appears to be quite complex in murine models of infarction or

ischemia/reperfusion. In vivo molecular imaging utilizing transgenic mice expressing Luciferase

Page 37: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

22

under an NF-κB promoter site revealed that post infarction NF-κB is up regulated with a peak

occurring at day 3. After that, the activity level decreases but remains elevated for up to 8 weeks

post-infarct (the maximum length these mice were followed) [85]. On one hand, NF-κB

activation is necessary to generate the cardioprotective response associated with ischemic pre-

conditioning, as several groups have demonstrated that this beneficial effect is lost with

inhibition of NF-κB signaling [72, 86-88]. On the other hand inhibiting NF-κB activation by

using p50 knockout mice [89], over-expressing A20 (an inhibitor of NF-κB protein) [90],

adenoviral delivery of IκB [91-92], or adenoviral delivery of MyD88 [93] led to decrease in

infarct size in rodent models of ischemia-reperfusion or infarction. Double-stranded

oligodeoxynucleotides with specific affinity for NF-κB used as decoys in rat hearts led to more

rapid recovery of heart function after heart transplantation [94] and reduced infarct size in a

murine model of ischemia-reperfusion [95]. In an isolated report, transgenic mice harboring a

mutated form of IκBα had increased susceptibility to tissue injury after acute left anterior

descending occlusion [96]. In addition small molecular inhibitors of NF-κB have shown

beneficial effects in reducing infarct size in murine models [97-101]. Interestingly one group of

researchers showed curcumin as having effective NF-κB inhibitory actions and ability to

attenuate plasma inflammatory cytokine surge and cardiomyocyte apoptosis following cardiac

ischemia-reperfusion [102-103]. These studies and the models they used are summarized below

in Table 2.

Page 38: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

23

Table 2. Summary of NF-kappa B studies with cardiac ischemia-reperfusion injury or myocardial

infarction.

Authors Journal/Year Model Infarct Size

Timmers, L et. Al.

[104]

Circ Res/2009 p50 KO/MI/mouse ↔/Adverse

Remodeling

Kawano, S et. Al.

[3]

AJPHCP/2006 p50 KO/MI/mouse ↔/Improved

Remodeling

Wakatsuki, S et. Al.

[105]

Expert Opin Ther/2008 IMD-0560/MI/rat ↔/Improved

Remodeling

Frantz, S et. Al.

[106]

FASEB J/2006 p50 KO/MI/mice ↔/Improved

Remodeling

Misra, A et. Al. [96] Circ/2003 mut IκBα/MI/mice ↑ Infarct Size

Meili-Butz, S et. Al.

[97]

Eur j Pharm/2008 Dimethyl fumarate/rat/IR ↓Infarct Size

Moss, NC et. Al.

[98]

AJPHCP/2007 Bay65-1942/mice/IR ↓ Infarct Size

Frantz, S et. Al. [89] Am J Pathol/2007 p50 KO/mice/IR ↓ Infarct Size

Trescher, K et. Al.

[91]

Cardiovasc Res/2006 ↑IκB expression/rat/MI Improved

Remodeling

Hua, F et. Al. [93] Bioch Biop Res /2005 Ad5-dnMyD88/mice/IR ↓ Infarct Size

Yeh, CH et. Al.

[102]

J Surg Res/2005 Curcumin/rabbit/IR ↓Infarct Size

Trescher, K et. Al. Eur J Cardioth Surg/2004 Ad-IκBα ↔/Improved

Page 39: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

24

[92] Remodeling

Onai, Y et. Al. [99] Cardiovasc Res/2004 IMD-0354/rat/IR ↓/Improved

Remodeling

Squadrito, F et. Al.

[107]

Lab Invest/2003 rAAV/IκBα/mice/IR ↓ Infarct Size

Pye, J et. Al. [100] AJPHCP/2003 20S proteasome inh/pig/IR ↓ Infarct Size

Kupatt, C et. Al.

[108]

Gene Ther/2002 NF-κB decoy/pig/IR ↓/Improved

Remodeling

Morishita, R et. Al.

[95]

Nat Med/1997 NF-κB decoy/pig/IR ↓ Infarct Size

None of these studies were designed to differentiate between inhibition of basal NF-κB

activity versus inhibition of surge in activity associated with ischemic damage and cytokine

release. With this background in mind, we hypothesized that blockade of cytokine mediated

surge in NF-κB occurring via the canonical or classical pathway with the NBD peptide, in

association with a PTD, would reduce infarct size in a mouse model of myocardial infarction.

Table 2 - continued

Page 40: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

25

2.0 PHAGE DISPLAY TO TARGET THE HEART

It is clear from the preceding chapter that protein transduction technology is rapidly evolving and

expanding in its applications, even though the exact mechanism of transduction remains

incompletely elucidated, and is unlikely to be the same for the different PTDs. The one limitation

these general PTDs have is their non-tissue specificity and ability to transduce a wide-range of

cell types limiting therapeutic efficacy and potentially increasing toxicity. Biopanning or phage

display would then be one useful technique to identify new PTD(s) which are tissue selective.

We therefore undertook phage display to screen a large peptide-display M13 phage library with

the goal of identifying peptide(s) able to transduce heart tissue selectively in vivo.

2.1 PHAGE DISPLAY

We screened a 12-mer M13 phage peptide display library (NEB, E8110S) for possible cardio-

selective peptide(s). We utilized a combinatorial in vitro single cycle followed by 3 in vivo

biopanning cycles. The M13 phage display peptide library is based on a library of randomized

dodecapeptides fused to a minor coat protein (pIII) of M13 phage. The displayed peptide (12-

mer) is expressed at the N-terminus of pIII, i.e., the first residue of the mature protein is the first

randomized position. The peptide is followed by a short spacer (Gly-Gly-Gly-Ser) and then the

wild-type pIII sequence. The library consists of approximately 2.7 x 109 electroporated

Page 41: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

26

sequences amplified once to yield approximately 100 copies of each sequence in 10 µl of the

supplied phage. Hence in the library there will be few phage that are relevant with a far excess of

irrelevant phage clones. To maximize selection of the relevant phage and minimize the large

pool of non-specific phage, the first cycle was carried out in vitro on H9C2 cells, a rat

cardiomyoblast cell line. These H9C2 cells have been characterized biochemically,

morphologically as well as electrophysiologically [109]. Although these cells showed similar

sugar residues on the surface coat as isolated rat cardiomyocytes, heart-specific morphological

structures were not detected. These cells did show outwardly rectifying, transient K+ current as

well as inward current through Ca2+ channels. Nevertheless, despite these differences, these

cells have been widely utilized to study molecular pathways involved in ischemia-related

apoptosis, hypertrophy as well as doxorubicin-mediated cardiac toxicity [110-114].

Rat cardiomyoblast cell line, H9C2 cells, (ATCC, CRL-1446), were passaged at least for

4 cycles. These cells were trypsinized and re-plated once they were ~70-80% confluent. After 4

cycles they were grown again to ~70% confluence in a 6-well plate and incubated with 10ul, or

~1011 pfu phage, for 6 hours, at 37˚C/5% CO2. At the end of the incubation period, the cells were

washed 6 times with pre-warmed PBS, trypsinized with 0.25% Trypsin EDTA (GIBCO/BRL)

for 5 minutes, and the cells pelleted by centrifugation for 5 minutes at a speed of 500rpms. The

cells were lysed by a single freeze-thaw cycle, and the internalized phage tittered by infecting

and growing in the provided E. coli bacteria. The recovered phage was amplified and re-tittered

before use in mice for in vivo biopanning.

For in vivo phage display, Balb/c, female, 10-12 week old mice were pre-treated with

intra-peritoneal chloroquine at a dose of 20mg/Kg the day before and the day of phage injection.

This is in contrast to prior reports of in vivo phage display, where short circulation times

Page 42: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

27

(typically 10-60 minutes) were used, since the purpose was to target the vascular endothelium of

various organs and tumors of interest. We utilized a prolonged incubation time as our aim was to

go beyond the endothelium and target cardiomyocytes. Molenaar et. Al. had reported that the in

vivo half-life of the M13 phage was 4.5 hours [115]. Since we aimed to recover actual,

internalized phage and reduce contamination from the pool of circulating phage, we employed a

much longer circulation time of 24 hours, equivalent to 5.5 half lives. In order to reduce

intracellular breakdown of internalized phage, we pre-treated mice with Chloroquine the day

before and day of phage injection (10mg/Kg, intraperitoneally), an agent shown to increase intra-

lysosomal pH in an attempt to increase chances of recovering internalized phage [116]. Phage

were injected intravenously (retro-orbitally), mice euthanized 24 hours later, heart and kidneys

dissected out, weighed and put through a collagenase digestion step. The isolated cells were

lysed with a single freeze-thaw cycle, recovered phage tittered, amplified, re-tittered and injected

again for a total of three in vivo phage display cycles. This experiment is represented

schematically in the figure (Figure 7) below. The reagents utilized for these experiments, as well

as the exact protocol details are presented in a step by step manner in Chapter 5, section 5.1:

Protocol for Peptide Phage Display.

Page 43: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

28

Euthanize, isolate myocytes, digest kidney, titrate recovered phage,

amplify phage, titrate amplified phage

Heart-target Kidney-control

Inject amplified phage i.v, circulate for 24hrs

+Isolate cell, amplify and titrate Phage

3 Cycles

Pre-treat mouse with i.p Chloroquine (20mg/Kg) 24hrs before and day of injection

H9C2 Cells Phage Library

Figure 7. Experimental design of the combinatorial in vitro and in vivo phage display.

The recovered phage was normalized by gram of tissue digested and expressed as a ratio

of heart to kidney and as an output to input ratio. The number of phage injected and recovered

with each cycle is presented in Table 3 below.

Page 44: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

29

Table 3. Number of phage injected and recovered from each cycle of phage display

4.4360.00000025.55E+012.46E+029.98E+084

0.7670.00000351.55E+031.19E+033.42E+083

0.0230.00001332.04E+074.63E+053.48E+102

0.00005955.95E+061.00E+111

Heart/Kidney RatioOutput/Input RatioOutput-KidneyOutput-Heart

Input PhageRound

These results are represented graphically in the figure below (Figure 8) showing heart to

kidney ratios (a.) and output to input ratios (b.).

Page 45: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

30

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

5

0 0.5 1 1.5 2 2.5 3 3.5 4 4.5

Biopanning Rounds

Hea

rt/K

idne

y R

atio

0

0.00001

0.00002

0.00003

0.00004

0.00005

0.00006

0.00007

0 0.5 1 1.5 2 2.5 3 3.5 4 4.5

Biopanning Rounds

Out

put/I

nput

Rat

io

b). Output to input ratio

Figure 8. Recovered phage normalized to tissue weight and plotted as a ratio of heart to kidney (a)

and output to input ratios (b).

a). Heart to kidney ratio

Page 46: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

31

Although the recovered number of phage and the output-to-input ratio decreased over

subsequent biopanning rounds, the ratio of heart-to-kidney phage increased with an exponential

increase from 3rd to the 4th cycle. The number of recovered phage decreasing over time is not

surprising as the viability is likely to decrease with repeat freeze-thaw cycles and storage over

time.

After the 4th round of biopanning, 10 plaques were picked and the inserted sequences

elucidated using the primers provided by New England Biolabs. Of these 10 plaques, 6 (60%)

were identical and carried the sequence gcgccgtggcatctttcgtcgcagtattctcgtact. This translates into

the following peptide sequence: Ala-Pro-Trp-His-Leu-Ser-Ser-Gln-Tyr-Ser-Arg-Thr

(APWHLSSQYSRT). This peptide was termed Cardiac Targeting Peptide or CTP and

synthesized by the peptide synthesis facility either conjugated to 6-carboxyfluoroscein,

biotinylated at the N-terminus, or as a combination peptide carrying the nemo-binding domain

peptide (NBD; sequence TALDWSWLQTE), which has been shown to inhibit activation of the

inducible NF-kappa B Kinase (IKK) by binding to the regulatory subunit (Nemo) of IKK.

Peptides were synthesized by Fmoc (N-(9-fluorenyl)methoxycarbonyl) solid phase

synthesis, N-terminally biotinylated or labeled with 6-carboxyfluoroscein, purified by reversed-

phase high performance liquid chromatography to >90% purity on an acetonitrile/H2O-

trifluoroacetic acid gradient, and confirmed by electrospray ionization mass spectrometry

(Peptide Synthesis Facility, University of Pittsburgh). Lyophilized peptides were reconstituted in

PBS to a 1 mM stock solution concentration. In some peptide preparations, due to variability in

the salt concentration, the peptide would not go completely into solution in PBS. In those

instances, a 40mM stock solution was made in DMSO, with subsequent dilution to 1mM

working solution by addition of PBS. Care was taken to use peptides suspended only in PBS or

Page 47: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

32

dH2O for use in cell culture, and minimal DMSO used to re-suspend peptides for use in animal

injections. All peptides were stored at -20˚C as a lyophilized powder, and re-constituted on the

day of use.

Calculating the physical properties of this peptide sequence CTP, using online tools,

reveals a molecular weight of 1432.6, a net charge of 1.1 at pH of 7.0, an isoelectric point

occurring at a pH of 9.35, with an average hydrophilicity index of -0.4

(https://www.genscript.com/ssl-bin/site2/peptide_calculation.cgi).

MW:

1432.55

PI:

9.35

Hydrophilicity Analysis:

Peptide Charge Attribute

APWHLSSQYSRT 2 Basic Note:

• Red: acidic residues, like D E and C-terminal -COOH

• Blue: basic residues, like R K H and N-terminal -NH2

• Green: hydrophobic uncharged residues, like F I L M V W and Y

• Black: other residues, like G A S T C N Q and P

Page 48: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

33

• Z: Unrecognized codes are replaced of 'Z'.

Figure 9. Physical properties of Cardiac Targeting Peptide or CTP.

In contrast to the earlier reported PTDs, like TAT or 16 amino acids from the third helix

of Drosophila Antennapedia homeodomain (penetratin), which are rich in cationic, basic amino

acids like arginine and lysine, this peptide sequence has only one arginine residue and is near-

neutral at physiological pH.

2.2 In vitro Transduction Experiments: Confocal Studies

In order to determine the ability of CTP to transduce cells in vitro, multiple different cell types,

along with H9C2 cells were incubated with varying concentrations of 6-carboxyfluoroscein

labeled CTP (CTP-6CF). A cardiomyoblast cell line (H9C2), a fibroblast cell line (NIH/3T3), a

murine fibrosarcoma cell line (MCA205), a cervical cancer cell line (HeLa) and a human tubular

cell line (HK-2), were passaged for a minimum of 3 cycles. After three cycles, cells were split

and plated onto collagen-coated cover-slips (H9C2, HK-2 cells) or regular cover-slips (NIH/3T3,

MCA205, HeLa cells) at a cell density of 2*105/well in a 6-well cell-culture plate. 24-hours after

plating, the media was aspirated, cells washed with 1ml of pre-warmed (to 37˚C) Optimem once

and left in 1ml of Optimem. Cells were then incubated at 37̊C/5%CO 2, with increasing

Page 49: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

34

concentrations (50uM, 100uM and 200uM) of CTP-6CF (1mM working solution in PBS) for 30

minutes, at the end of which cells were washed 6 times with pre-warmed PBS, fixed with 2%

paraformaldehyde (see Chapter 5, Methods, Section 5.2.1 for “cell transduction experiments with

CTP-6CF, and section 5.2.2 for details on making 8% paraformaldehyde stock solution) for 30

minutes to which 1ul of a 1000X DRAQ5 (Molecular Probes, F1303), a nuclear stain, was

added. After 30 minutes, PFA was aspirated, cells washed with pre-warmed PBS once, and the

cover-slips mounted onto glass slides using gelvitol. After setting overnight, at 4˚C, light-

protected, cells were examined by confocal fluorescent microscopy. Below are confocal

micrographs of various cell types showing preferential transduction of H9C2 cells over other cell

types (Figure 10).

Page 50: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

35

3

3T3

No Rx 50uM 100uM 200uM

HeLa

HK-2

H9C2

MCA205

Figure 10. Confocal micrographs of H9C2, 3T3, MCA205, HeLa and HK-2 cells incubated with

increasing concentrations of CTP-6CF; Nuclei - blue (DRAQ5), CTP-6CF - green, 20x. Scale bars represent

100uM.

Page 51: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

36

These micrographs show uptake of CTP-6CF by H9C2 cells in a dose-dependent fashion

with increasing uptake at higher concentrations. Uptake by 3T3, MCA205 and HeLa cells occurs

to a much lesser extent and only at the highest end of the dose used (200uM, last panel). No

uptake at all was seen at any of the tested doses by HK-2 cells, a human renal tubular cell line.

Transduction experiments with H9C2 cell line were repeated with 200uM of CTP-6CF,

incubated for 30 and 60 minutes and higher magnification confocal micrographs obtained. As

shown in the figure below (Figure 11), CTP-6CF is intracellular with a punctuate distribution,

similar to what has been reported for TAT, suggesting perhaps an endosomal localization of the

peptide after internalization.

Page 52: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

37

Figure 11. High magnification confocal micrographs of H9C2 cells incubated with CTP-6CF for 30

(top row) and 60 minutes (bottom row).

There has been concern voiced in literature following the reports on cationic cell

penetrating peptides (CPP) that transduction with nuclear localization might be a result of a

fixation artifacts [117-118]. TAT, a cationic, basic PTD, because of large number of arginine and

lysine residues, has been shown to bind strongly to surface glycoproteins especially heparan

H9C2 cells incubated with CTP-6CF for 30 mins: 40x and 60x

H9C2 cells incubated with CTP-6CF for 60 mins: 40x and 60x

Page 53: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

38

sulfate. This binding has been shown to be followed by endocytosis leading to internalization

[119]. Thus the concern has been that transduction may simply be binding of positively charged

peptides to heparan sulfate followed by endocytosis and any cytosolic or nuclear localization is a

fixation artifact. To confirm that the above findings are actual transduction and not simply a

fixation artifact, we followed H9C2 cells incubated with a random peptide sequence labeled with

6CF (RAN-6CF), CTP-6CF or TAT-6CF with live imaging without any fixation protocols. The

random peptide (ARPLEHGSDKAT) was the sequence of a phage picked from the original

library that had not undergone any selection pressures of phage display. The figure below

(Figure 12) presents the results after 60 minutes of incubation in which a DIC image is over-laid

on the fluorescent image to give cell structural detail.

RAN-6CF-200uM CTP-6CF-200uM

TAT-NBD-6CF-50uM

Figure 12. Live cell imaging of H9C2 cells incubated with RAN-6CF, CTP-6CF or TAT-6CF.

Page 54: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

39

These micrographs show intracellular and intranuclear fluorescence with both TAT-6CF

and CTP-6CF, with no fluorescence uptake with RAN-6CF. To assess transduction without

fixing cells or using fluorescent microscopy, we also did a functional assay to prove

transduction, as detailed below.

2.3 IN VITRO TRANSDUCTION: FUNCTIONAL ASSAY

In order to assess transduction of H9C2 cells with CTP peptide, we used a reporter-Luciferase

based cell assay, as described by Madge and May [120]. H9C2 cells were plated onto 96-well

plates at a cell density of 2*104cells/well. Next day cells were transfected with a reporter plasmid

expressing Luciferase under an NF-κB binding promoter site and a control Renilla expressing

plasmid, using Lipofectamine. Twenty-four hours post-transfection, cells were pre-treated with

increasing concentrations of the CTP-NBD (nemo-binding domain) peptide. NBD binds to the

nemo binding site of IKKβ and to a lesser extent IKKα, preventing its association with NEMO or

IKKγ, thus preventing activation of the IKK complex. This prevents phosphorylation of IκB and

its subsequent degradation and release of NF-κB, which is not activated and remains cytosolic.

Cells pre-treated with CTP-NBD were then challenged with TNFα, an NF-κB activator, and

Luciferase activity measured in cell lysate. The methodological details of these set of

experiments are provided in Chapter 5, Methods, section 5.3: Methodology for transfection of

cells and inhibition of NF-κB activation. Figure 13 gives a schematic presentation of the

experimental protocol.

Page 55: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

40

Split/Plate H9C2 Cells

Transfect cells with NF-kappa B/Luciferase plasmid

Pre-treat cells with CTP-NBD for 30 mins

Stimulate with mTNFα(10ng/ml) for 3 hrs

Lyse, collect cells, do Luciferase assay on the supernatant

Figure 13. Experimental protocol for the in vitro transduction experiments using Luciferase readout.

All work was done in quadruplicate. The Luciferase readings were corrected for

transfection efficiency differences using the Renilla readout. Differences across groups were

compared using a two-tailed Student’s t-test. A p-value of <0.05 was considered statistically

significant. Below are the results of experiments carried on H92 cells.

Page 56: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

41

0

0.5

1

1.5

2

2.5

3

3.5

4

No Rx Plasmid TNFα CTP-NBD-50 CTP-NBD-100 CTP-NBD-200 CTP-NBD-400

Treatment Groups

Fold

NF-

kapp

a B

Act

ivat

ion

Figure 14. Inhibition of Luciferase activity by CTP-NBD in H9C2 cells.

The exact same experiment with identical conditions was repeated in MCA205 cell line

(Figure 15) as well as in HeLa cells (Figure 16).

Page 57: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

42

Figure 15. Lack of inhibition of Luciferase activity by CTP-NBD in MCA205 cells.

CTP-NBD was not able to inhibit TNFα mediated activation of NF-κB in MCA205 cells

even at the highest tested dose of 400uM.

Page 58: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

43

Figure 16. Relative lack of inhibition of Luciferase activity by CTP-NBD in HeLa cells.

CTP-NBD failed to inhibit TNFα mediated NF-κB activation, except at the very highest

dose used (400uM). This is consistent with the confocal micrograph data where transduction of

HeLa cells by CTP-6CF occurred marginally at the highest dose used of 200uM (Figure 10).

Following these inhibition of NF-κB activation studies, we performed cell viability

studies to assess changes related to peptide treatment. The experiment was repeated with H9C2

cells being doubly-transfected and pre-treated with CTP-NBD in increasing concentrations,

followed by TNFα (10ng/ml) challenge for 3 hours. After this time period, the media was

aspirated and replaced with MTT assay media to assess for cell viability. As shown below,

inhibition of NF-κB with CTP-NBD led to decrease in cell viability in a CTP-NBD dose-

0

50

100

150

200

250

300

350

400

450

No Rx Plasmid TNFα CTP-NBD-50 CTP-NBD-100 CTP-NBD-200 CTP-NBD-400

Treatment Groups

Corrected Luciferase Activity

Page 59: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

44

dependent fashion. All treatment groups were done in quadruplicate. Error bars represent 1

standard deviation (SD).

Figure 17. H9C2 cell viability after transfection, treatment with CTP-NBD and subsequent TNF-

alpha treatment.

This finding is consistent with reports in the literature that NF-κB inhibition increases

apoptosis in H9C2 cells challenged with TNFα [121]. These results clearly demonstrate a

biological effect of the NBD peptide that can only occur if the peptide was internalized by the

PTD action of CTP. This decrease in cell viability did not occur with TNFα treatment or CTP-

NBD treatment alone as shown in Chapter 4, Figure 40.

In summary, in this section we have shown transduction occurring preferentially of H9C2

cells, a rat cardiomyoblast cell line, over a fibroblast, a fibrosarcoma, a cervical epithelial cancer

cell line and human tubular kidney cell line using confocal fluorescent microscopy. We have also

Page 60: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

45

demonstrated preferential transduction of H9C2 cell line over MCA205 cells or HeLa cells using

a reporter plasmid carrying Luciferase expression under the control of an NF-κB binding

promoter site. Next we will show transduction of heart tissue occurring in vivo with fluorescently

labeled CTP.

2.4 IN VIVO TRANSDUCTION STUDIES

Our in vitro data showed preferential targeting of H9C2 cells over other, tested cell lines as

assessed by confocal imaging, as well as a functional assay using Luciferase as a readout. This

naturally led to the next question of whether preferential targeting of heart tissue will occur in

vivo as well. The initial in vivo targeting studies were performed using CTP-6CF. Female

Balb/C mice were anesthetized using 2.5% Avertin, 12-15ul/gm of body weight, injected

intraperitoneally. After adequate anesthetic levels were achieved, as assessed by lack of limb

withdrawal to toe pinch or lack of a corneal reflex, mice were injected retro-orbitally with CTP-

6CF (25 mg/Kg) and euthanized 15 minutes later. The heart was dissected out, washed in PBS,

placed in 2% paraformaldehyde (PFA) for 4 hours at room temperature (RT), followed by 30%

sucrose, 4ºC, over night. Next day hearts were flash frozen in liquid nitrogen-chilled Isopentane

and placed in -80ºC for later cryosectioning. After embedding in OCT, hearts were cryosectioned

in the short-axis as 6uM thick sections and stained with DRAQ5, a nuclear stain. The sections

were also stained for actin using phalloidin Alexa- 647 (Molecular Probes, A22287) and stained

for laminin using a rabbit anti-laminin antibody followed by a goat anti-rabbit Cy3 (Jackson

ImmunoResearch, 111-167-003) secondary antibody. Confocal microscopy was performed on

these heart sections. The figure below (Figure 18) illustrates a section showing intracellular

Page 61: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

46

localization of CTP-6CF, co-localizing with Actin. Laminin, which is present only on the surface

of cells and hence has a ring-like appearance around myocytes, does not co-localize with CTP-

6CF, which has a distinctly different distribution pattern.

Figure 18. Transduction of mouse heart by intravenously injected CTP-6CF at 15 minutes.

Female, Balb/C mice were injected retro-orbitally with PBS or CTP-6CF at a dose of

25mg/Kg, euthanized at 15 minutes, hearts dissected and treated as outlined above. Five non-

overlapping, contiguous sections were taken from each heart for quantification of green

fluorescence (CTP-6CF) expressed as a percentage of total area (blue; stained for actin). There

were 3 mice in each group, and 5 sections quantified from each heart and averaged.

The figure below (Figure 19) shows the mean amount of fluorescence in all sections. The

error bars represent one standard error of the mean.

CTP-6CF Actin

Laminin Merged

Page 62: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

47

2

4

6

8

10

12

14

16

18

20

CTP PBS

Treatment Groups

Perc

ent A

rea

of F

luor

esce

nce

Figure 19. Quantification of fluorescence in mouse hearts (n=3 in each group).

Next the biotinylated form of CTP (200uM) or PBS was incubated with Streptavidin-

Alexa488 conjugate (10ul of a 2mg/ml solution), at RT, light-protected, for 60 minutes, prior to

injection intravenously (retro-orbitally) into Balb/C, female mice. An equivalent volume of

PBS+streptavidin-alexa488 was injected as a negative control. Mice were euthanized at 30

minutes, heart, liver, kidney, lung, skeletal muscle (hind-limb quadriceps), brain and spleen

dissected out and treated as outlined above. The sections were stained with DRAQ5, cover-

slipped and confocal microscopy performed. For confocal microscopy, laser intensities/gains

were set using negative control (PBS injected) heart tissue to minimize background fluorescence.

Once laser intensity for the negative control hearts was set, it was kept constant across all

subsequent imaging. Also serial scanning was performed to prevent “bleed-through” from one

laser wavelength to another. Figure 20 below illustrates robust uptake of the peptide, as judged

Page 63: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

48

by tissue fluorescence, with very little uptake by lung and kidney glomerular capillaries and none

by skeletal muscle, brain, liver or spleen (data for spleen not shown).

Page 64: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

49

Skeletal Muscle

Liver

Kidney

Lung

Brain

Figure 20. Transduction of heart tissue after intravenous injection with CTP-biotin-streptavidin-Alexa488

complex.

CTP+SA488 PBS+SA488

Page 65: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

50

Balb/C, female mice were also injected with the CTP-biotin-streptavidin-Alexa488

conjugates and euthanized at different time points to study the biodistribution of this complex.

The figure below, (Figure 21), shows transduction beginning at 15 minutes, localized to the sub-

epicardial region, with more diffuse uptake at 30 minutes and almost complete absence of

fluorescence at 120 minutes.

Figure 21. Biodistribution of CTP-biotin-streptavidin-Alexa488 complex after intravenous injection

Heart Kidney Liver

15mins

30mins

60mins

Page 66: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

51

A random peptide (RAN; ARPLEHGSDKAT), picked from the original, unselected M13 phage

library, was synthesized in a biotinylated form. Biotinylated RAN or CTP peptides were labeled

with neutravidin-conjugated fluospheres (Molecular Probes, F8770) with an overnight incubation

at 4ºC. These fluospheres are 40nm in diameter and fluoresce at an excitation wavelength of 605

nm, allowing for in vivo bead tracking. Female Balb/C mice received intracardiac injections of

fluospheres-labeled RAN peptide, CTP peptide or control PBS incubated with fluospheres alone.

Mice were anesthetized with isoflurane delivered by the XGI-8 Gas Anesthesia System

(Xenogen). Initial isoflurane concentration was set to 2.5% and was reduced to 1.5% once the

animal was anesthetized. Mice were then imaged at 30, 60, 120, and 180 minutes post-injection

with the IVIS Lumina (Caliper Life Sciences Inc.). The figure below (Figure 22) shows

localization of fluorescence to the heart in mice injected with CTP+fluospheres, with rapid

dissipation of fluorescence after injection with RAN+fluospheres or PBS+fluospheres. After

three hours of imaging, these mice were euthanized, hearts dissected, processed as detailed

above, cryosectioned, cross-stained with DRAQ5 and confocal microscopy performed.

Page 67: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

52

Figure 22. In vivo, whole mouse imaging after intra-cardiac injection of CTP, RAN or PBS

conjugated to fluorescent 40nm beads.

30mins

60mins

120mins

180mins

Fluospheres Only CTP+Fluospheres RAN+Fluospheres

Page 68: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

53

The confocal micrographs below (Figure 23) show a few myocytes with uptake of the

fluospheres, actually much less than the whole mouse, in vivo imaging would suggest. Confocal

microscopy did show fluorescent beads sticking to the endocardial region suggesting that these

beads might only be able to stick to the surface and not be internalized as readily as CTP-6CF or

CTP-biotin-streptavidin-alexa488 complexes. These beads are 40nm in size and the association

with CTP-biotin may not necessarily be a one-to-one association leading to larger complex

formations. This is indirectly supported by the observation that the solution injected was a highly

viscous one and required a large-bore, 18G needle, precluding a peripheral tail-vein or retro-

orbital injection.

Figure 23. Confocal micrographs of hearts injected with PBS+fluospheres, RAN+fluospheres or

CTP+fluospheres.

To demonstrate cell specificity of the CTP peptide in vivo, we undertook a second set of

experiments. Balb/C, female mice were anesthetized with intra-peritoneal Avertin (2.5%, 10-

Page 69: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

54

15ul/gm of body weight), intubated with a 22G cannula using direct laryngoscopy and

visualization of the vocal chords, and placed on a rodent ventilator (Harvard apparatus). The

chest cavity was entered using a left, lateral ternotomy approach, and an anterior infarct

produced by placing a suture through the anterior wall, 2mm inferior to the lowest dip of the left

atrial appendage to blindly ligate the left anterior artery. Infarct was confirmed by the resulting

pallor of the anterior wall and apex as well as wall motion abnormality produced in the region.

The chest wall was closed in two layers, and the animal placed on a heating pad until awake. One

week post-infarct, mice were re-anesthetized and injected intravenously with CTP-biotin-

Streptavidin-Alexa488 (SA488) complex or an equivalent volume of PBS with SA488 (10mg/Kg

body weight). After 30 minutes of circulation time, mice were euthanized, hearts dissected out,

washed, fixed, cryosectioned, cross-stained with DRAQ5, and confocal microscopy performed.

Contiguous sections were also H&E stained to show the infarct area. Figure 24 below shows

uptake of the fluorescently labeled CTP by normal heart tissue with exclusion of uptake from the

infarcted, scarred myocardium (arrows). No uptake of PBS+SA488 alone was seen in any area of

the heart. This experiment demonstrates in vivo cell specificity of our peptide, CTP, as it is taken

up by normal myocardium and excluded from the scarred, infracted heart. The infarct is

demonstrated in the H&E section by the lighter pink, thinned out area.

Page 70: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

55

Figure 24. Uptake of CTP-6CF preferentially by normal myocardium with exclusion from the infarct

area.

Our lab has shown that a homopolymer of lysine, 8K, is able to transduce multiple cell

types effectively and consistently exceeding those of TAT and arginine homopolymers [16]. In

order to compare transduction of heart tissue by CTP versus 8K, biotinylated form of these two

peptides as well as a random peptide (RAN; mentioned above) were incubated with Streptavidin-

Alexa488 (SA488) for 60 minutes. Balb/C, female mice were anesthetized with intra-peritoneal

Avertin and injected intravenously with CTP-SA488, 8K-SA488, or RAN-SA488 at a dose of

10mg/Kg. At the end of a 30 minute circulation time, mice were euthanized, hearts and other

organs dissected and processed as detailed above for cryosectioning and subsequent confocal

microscopy. The figure below (Figure 25) shows transduction of heart tissue by CTP-SA488 in a

Page 71: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

56

generalized manner at 30 minutes, with little uptake by kidney capillaries and none by the liver.

In contrast 8K demonstrates robust transduction of kidney glomeruli and liver tissue, with little

uptake by the heart. In contrast RAN-SA488 is not taken up by any of the tissues tested.

Figure 25. Comparison of heart tissue transduction between CTP, 8K and RAN peptide.

Heart

Kidney

Spleen

CTP+SA488 8-Lysine+SA488 Random+SA488

Liver

Page 72: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

57

2.5 HUMAN HEART TISSUE EXPERIMENTS

Our work above has shown that in vivo phage display can be used to screen large phage display

libraries for peptide(s) able to transduce cardiomyoblasts cell lines and heart tissue preferentially

in vivo after a systemic, peripheral intravenous injection. The identification of such a peptide,

CTP, raises many interesting and relevant questions. One key one would be whether this cardiac

targeting is species specific or not. If such was the case, the utility of this peptide would be

limited. In order to test whether CTP is able to transducer human heart tissue, we performed

experiments ex vivo on explanted heart tissue.

Heart transplantation is a destination, life-saving therapy for end-stage congestive heart

failure. At the University of Pittsburgh Medical Center (UPMC) anywhere from 30 to 50 heart

transplants are performed in a year. The Institutional Review Board has approved a protocol

whereby patients awaiting heart transplant can consent to the use of explanted, diseased hearts

for scientific experimentation with appropriate measures taken to protect patient identification

and privacy. An extension of this protocol was sought where I could share tissue obtained at the

time of heart transplant, in a blinded fashion, to run ex vivo incubation experiments with various

fluorescently labeled peptides, including CTP.

Human explanted hearts were obtained from the operating rooms of UPMC at the time of

ongoing heart transplants. Heart tissue was dissected from the proximal part or the base of the

heart to avoid the more scarred, infarcted, distal tissue. Small 1-2mm thick slices of heart tissue

were taken and incubated with CTP-6CF (500ul of a 1mM solution) or PBS, for 30 and 60mins,

at 37ºC / 5% CO2. At the end of the incubation period, the heart slices were washed in PBS 6

times, fixed in 2% paraformaldehyde for 4 hrs at room temperature, followed by 30% sucrose

overnight at 4ºC. Next day the tissue was frozen in liquid nitrogen-chilled isopentane and stored

Page 73: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

58

at -80ºC, for later cryosectioning, cross-staining with DRAQ5 and confocal microscopy.

Confocal micrographs were obtained avoiding the edges because of the necessary damage

associated with dissecting the heart tissue pieces. Confocal micrographs were taken 3-4 cell

layers away from any edges. The figure below (Figure 26) demonstrates internalization of the

peptide into heart myocytes.

Figure 26. Transduction of human heart tissue ex vivo by CTP-6CF.

PBS CTP-6CF-30mins

CTP-6CF – 60mins; 40x and 60x

50uM

Page 74: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

59

To investigate if this transduction was indeed peptide specific, and not secondary to

increased membrane leakiness resulting from tissue being ischemic, we incubated thin, 1-2mm

thick, heart slices, from a separate explanted heart, in only 6CF in PBS, CTP-6CF, RAN-6CF as

well as 6CF labeled 6-Arginine, a known PTD. We also incubated heart slices with Evans blue

dye (EBD) to assess for membrane leakiness. Evans blue dye fluoresces with an excitation

wavelength of 620nm and emission wavelength of 680nm [122-123], making it possible to be

imaged with fluorescent microscopy using the appropriate lasers for excitation. Heart tissue was

incubated in 500ul of 1mM solution of all the peptides in PBS, for 30mins at 37̊C/5%CO 2. At

the end of the incubation period, heart slices were rinsed thoroughly 6 times in 1ml of PBS and

fixed in 2% paraformaldehyde for 4 hours, at room temperature, light-protected, followed by

30% sucrose, overnight at 4̊C. Next day tissue was frozen in liquid nitrogen chilled Isopentane,

embedded in OCT, cryosectioned, cross-stained with DRAQ5, mounted and confocal

microscopy performed as detailed above. Confocal microscopy showed robust and diffuse uptake

of both 6R-6CF and CTP-6CF, with occasional cells showing fluorescence in RAN-treated or

6CF-treated hearts. Confocal microscopy of the EBD treated hearts did not show any

fluorescence indicative of uptake which would have occurred had the membranes been

significantly damaged by length of ischemia. The figure below (Figure 27) shows confocal

micrographs of heart tissue treated with different peptides or controls.

Page 75: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

60

PBS CTP-6CF 6CF

6R-6CF CON-6CF EBD

Figure 27. Results of ex vivo human heart tissue experiment at 30 mins.

The result of these ex vivo experiments demonstrates that CTP-6CF is indeed internalized

by human myocytes as demonstrated by confocal microscopy and is not simply sticking to the

outer surface. Moreover, this internalization is not seen with simply 6CF, the fluorescent

chemical used to label all our synthesized peptides, and is not seen also with a random peptide.

In contrast, 6R-6CF (homopolymer of 6 arginine amino acid residues), a known PTD, is indeed

able to transducer these human myocytes ex vivo. Lastly, this internalization is not simply a

result of increased membrane leakiness due to ischemia, as evidenced by lack of Evans blue dye

uptake by the myocytes.

Page 76: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

61

2.6 CHAPTER SUMMARY

In this chapter we have presented data demonstrating that a combinatorial approach of cell

culture and in vivo biopanning or phage display can be utilized to identify a peptide, Cardiac

Targeting Peptide (CTP), of transduction potential. This peptide is able to transduce a rat

cardiomyoblast cell line, H9C2 cells, in vitro. This transduction is cell-specific and does not

occur at all, or occurs at very low levels, in mouse fibroblast cell line, a fibrosarcoma cell line,

human epithelial cancer cell line or human tubular endothelial cell line. We demonstrate that this

peptide is not simply sticking to the surface and being internalized as a result of a fixation

artifact with our functional Luciferase assay. In our assay Luciferase is expressed under the

control of an NF-κB promoter that requires NF-κB activation to lead to expression of the protein.

NF-κB is a cytoplasmic protein that acts as a transcription regulator and enters the nucleus after

activation and joining of its sub-units, the commonest being hetero- or homo-dimers of p50 and

p65 subunits. Our peptide is able to carry and internalize an 11-amino acid peptide, NBD (nemo-

binding domain), that inhibits this activation. In order to inhibit Luciferase expression, CTP must

be internalized and escape the endosomal compartment and deliver NBD to the cytoplasm where

NF-κB resides and prevent its subsequent activation.

We have gone on to demonstrate that this transduction is indeed occurring in vivo as

well, and is specific for the heart in mice. After a peripheral intravenous injection, CTP-6CF is

seen to transducer mouse hearts robustly with little uptake by liver or spleen, with only a few

lung and kidney capillaries showing fluorescence. This transduction peaks at 15mins for the

smaller CTP-6CF, a 1790 Da molecule, but occurs at 30mins for the larger CTP-biotin-

Streptavidin-Alexa488 (CTP-SA488), an ~55kDa in size complex. The biodistribution studies

revealed that transduction begins to occur as early as 15mins, peaks at 30mins and is almost gone

Page 77: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

62

by 120mins. At the same time, the fluorescence seems to increase in kidney glomerular capillary

cells, suggesting that this might be a route of excretion for the peptide or at least the fluorescent

marker, 6CF, after peptide breakdown.

Labeling the biotinylated form of our peptide CTP with red fluorescent streptavidin

conjugated fluospheres made it possible to track this peptide in vivo using whole mouse imaging.

CTP complexed with these 40nm fluospheres was injected intracardiac and mice followed by

whole mouse imaging over multiple time points. The fluorescence was seen localized to the heart

over 3 hours, as opposed to unconjugated beads or random peptide (RAN) conjugated to these

beads. To further demonstrate the in vivo specificity of this peptide for normal heart tissue, mice

underwent infarcts and a week post-infarct were injected with CTP-SA488 or PBS+SA488

complex. Confocal microscopy revealed fluorescence in normal heart tissue with complete

exclusion of fluorescence from the thinned-out, infarcted, scarred heart tissue, thus

demonstrating cell specificity in vivo.

In order to compare transduction efficiencies relating to the heart, mice were injected

with fluorescently labeled CTP, 8K or RAN. Confocal microscopy showed transduction of the

heart with CTP, with no significant heart transduction noted with 8K or RAN peptide, at similar

doses and identical time-points studied.

A peptide able to transduce heart tissue in vivo in mice alone would have limited

applicability in terms of developing therapeutics or new imaging agents of ultimate clinical use.

Therefore we sought to determine if CTP is able to transducer human cardiac myocytes ex vivo.

As per a pre-existing IRB protocol, explanted heart tissue was collected from consenting patients

undergoing heart transplants for end-stage congestive heart failure. A modification of this

protocol allowed us to have access to this tissue in a blinded fashion. Thin, 1-2mm, slices of

Page 78: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

63

freshly explanted human heart tissue were incubated with varying concentrations of CTP-6CF

for 30 mins. Tissue was washed thoroughly in PBS and fixed and cryosectioned for later

confocal microscopy. This revealed robust transduction of heart tissue, with minimal uptake of

just 6CF, RAN, and no uptake of Evans blue dye (a marker used to assess membrane

permeability). 6R, a known PTD used as a positive control in our experiments, was taken up by

cardiomyocytes robustly as well.

In conclusion, using a combinatorial approach of cell culture and in vivo phage display

we have identified a peptide able to transducer rat cardiomyoblasts in a cell-specific manner in

vitro, mouse heart tissue selectively in vivo and human heart tissue ex vivo.

Page 79: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

64

3.0 MURINE INFARCT SIZE STUDIES USING NBD

NF-κB was first identified as a transcription factor binding to the intronic enhancer of the Кappa

light chain gene in B-cells over 20 years ago [44-45]. Since that initial description interest and

research regarding this factor has been occurring at a blistering pace. As detailed in Section 1.6,

NF-κB is unregulated in response to ischemia-induced surge in cytokines, primarily TNF-α and

IL-6. Interestingly neutrophil depletion reduced this increase in TNF-α and IL-6, and reduced

nuclear translocation of NF-κB [124]. In section 1.4, we detailed the role of IKKγ or NEMO and

its interaction with the catalytic proteins IKKα and IKKβ. The specific domain responsible for

the interaction of NEMO with NBD has been worked out and termed the NEMO-binding domain

[52]. The same report also detailed out how a peptide, NBD peptide, complexed with

Anntenapedia homeodomain, a PTD, was able to reduce TNF-α mediated NF-κB activation

without diminishing the basal levels of NF-κB activity. In fact the basal NF-κB levels were

somewhat increased in HeLa cells treated with the peptide (two-fold) as compared with

untreated, control cells (40). Furthermore, NBD in association with varying PTDs (TAT or

Anntenapedia) has been demonstrated to decrease inflammation associated pathologies

(rheumatoid arthritis, inflammatory bowel disease) in animal models (see section 1.5 for more

details). We therefore hypothesized that inhibiting NF-κB activation in a murine model of

infarction by delivering NBD in association with a PTD at the time of ischemia would reduce

infarct size.

Page 80: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

65

3.1 GENERATION OF THE ANIMAL MODEL OF MYOCARDIAL INFARCTION

In order to test the above hypothesis, an animal model of myocardial infarction was necessary.

The following protocol was used and kept consistent from one set of experiments to another and

between groups. Female, Balb/c, 10-20 week old mice were used for these set of studies. Female

mice were chosen because of the ability to house 5/cage, as opposed to male mice (4/cage) and

more importantly because female mice are able to tolerate cardiac insults better with lower post-

operative mortality [125]. Mice were anesthetized with intraperitoneal (i.p.) injection of 2.5%

Avertin (a mixture of 15.5 ml tert-amyl alcohol to 25 grams of 2-2-2 Tribromoethanol - stock

solution diluted to 2.5% concentration with PBS) at a dose of 15ul/gm of body weight. Adequate

anesthetic level was assessed by lack of response to toe-pinch and lack of a gag reflex. Mice

were intubated, using direct visualization of the vocal chords, with a 22G cannula and placed on

a Harvard rodent ventilator apparatus. Tube placement was confirmed by symmetric expansion

of the thoracic cavity. Mice were ventilated with ~0.1cc/breath tidal volume at a rate of 110

breaths/minute.

Following successful intubation/ventilation, the chest cavity was opened using a left

lateral ternotomy approach. Care was taken not to extend the surgical incision cephalad to the

level of the sternal head, as a large venous confluence is present in that region and cutting

through them can lead to major, exsanguinating bleed. A self-retaining mouse retractor was

placed inside the incision and the teeth opened to reveal the anterior surface of the heart. The

pericardium was gently removed with blunt forceps. A suture, 6-0 prolene, blue, monofilament

(Owens & Minor: #8714H) was placed along the lateral wall of the left ventricle (LV), ~2mm

below the lowest dip of the left atrium and ligated with exactly 5 squarely-placed knots. Ischemia

was confirmed by the resulting pallor of the myocardium and development of wall motion

Page 81: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

66

abnormality. The muscle layer and skin were closed in two layers with running sutures with 8-0

black, monofilament nylon suture (Owens& Minor: #2808G). The mice were extubated once

spontaneous breathing was confirmed, and recovered on a heating pad. Post-operative pain was

managed with subcutaneous injection of Ketoprofen, 5mg/Kg, on post-op day 0, 1 and 2 or with

subcutaneous Buprenorphine, at a dose of 0.1mg/Kg body weight given once on the day of

surgery and twice daily on post-op day 1 and 2. Mice also got one injection of subcutaneous

Ampicillin, 100mg/Kg, after surgery for prophylaxis against post-surgical infection. Right after

ligation and suture placement on the anterior surface of the heart to produce myocardial

infarction, and before closing the chest wall, mice received i.p. peptides in different doses. The

surgeon (myself) was always blinded to the treatment. Also surgeries were carried out in a

rotational fashion so that any variation due to surgical technique was distributed equally over the

treatment groups.

Mice were euthanized on post-operative day 7. On day of euthanasia, mice were

anesthetized, chest cavity opened, and 1cc of 1M KCl injected via the right ventricle (RV) in

order to arrest the heart in diastole to prevent contraction band necrosis. This was followed by

injecting, through the right ventricle, 5cc of Glyo-fixx (formalin-substitute), heart dissected out

and placed in 15ml of Glyo-fixx, until tissue embedded for sectioning. After paraffin embedding,

cross-sections of the heart were taken starting at the apex. The first section was taken once the

LV cavity appeared, with second and third sections taken 10 microns apart and cephalad of the

first section. The three sections were H&E stained, light microscopy photographs taken, and area

of infarct in each section mapped out using computer software Metamorph and expressed as a

percent of the whole heart. The infarct area from the three sections was averaged to give the final

Page 82: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

67

infarct size for each mouse. All mouse protocols were approved by the University of Pittsburgh

Institutional Animal Care and Use Committee (IACUC).

3.2 INFARCT STUDIES USING 8K-NBD

In view of the above background and after establishing the murine infarct model, we set out to

test the following hypothesis. We hypothesized that suppression of NF-κB activation using the

NBD peptide, with 8K as a delivery PTD, would reduce infarct size in a mouse model of

myocardial infarction. Prior to the actual infarct study, we undertook whole mouse in vivo

imaging to show that 8K-NBD is able to suppress NF-κB activation in response to LPS

stimulation, a known activator of NF-κB.

To demonstrate that 8K-NBD is indeed able to suppress NF-κB stimulation, we used

Luciferase expressing transgenic mice with Luciferase expression controlled by an NF-κB

promoter site. Mice were injected intraperitoneally (i.p.) with 8K-NBD (10mg/Kg) at time 0 and

2 hrs with LPS injected immediately after the first 8K-NBD injection. Whole mouse in vivo

imaging for Luciferase activity was performed at 30mins, 2hrs, 4hrs and 6hrs. As illustrated in

the figure below, initial Luciferase signal was similar between the two groups of mice but at 4

and 6hrs the signal increased in the LPS injected mice and remained stable/suppressed in the 8K-

NBD treated mice.

Page 83: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

68

LPS 8K-NBD+LPS

Baseline

30 mins

4 hrs

2 hrs

Page 84: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

69

Figure 28. Inhibition of LPS induced Luciferase signal by 8K-NBD

After these initial imaging experiments, we proceeded to the infarct studies. Mice

underwent left artery ligation, as detailed above, and were treated with i.p. PBS, 8K-NBD or 8K-

mutant NBD (8K-mutNBD; KKKKKKKK-TTLDASALQME), at a dose of 5mg/Kg/injection,

on post-op day 0 (immediately post-ligation) and post-op day 1 and day 2. Treatments were

color-coded and administered in a rotational fashion. The surgeon was blinded to the treatment

groups. Mice were euthanized on post-op day 7, heart dissected out and infarct size calculated, as

detailed above in Section 3.1. A record of post-op mortality was kept. Also body weight and

heart weight were recorded on day of euthanasia. There was an n=6 in each group. Figure 29

below shows the post-op mortality in each of the three groups.

6 hrs

Page 85: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

70

Figure 29: Post-operative mortality by treatment group in the 8K-NBD study.

However the heart-weight to body-weight ratios did not differ across the three groups as

illustrated in Figure 30 below.

0

5

10

15

20

25

30

35

8K-NBD 8K-mutNBD PBS

Per

cent

Mor

talit

y

Treatment Groups

Surgical Mortality by Treatment Groups

Maliha
Placed Image
Page 86: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

71

Figure 30. Heart weight to body weight ratios across the treatment groups in the 8K-NBD study.

In contrast, and contrary to our expectations, the infarct size were significantly different

across groups and worse in mice treated with 8K-mutNBD (Figure 31). Mice treated with 8K-

NBD had larger infarct sizes than PBS treated mice, though the difference showed a trend

without reaching statistical significance.

0

1

2

3

4

5

6

7

8

8K-NBD 8K-mutNBD PBS

Rat

ios

(mg

/gm

)

Treatment Groups

Heart-Weight to Body-Weight Ratios

Maliha
Placed Image
Page 87: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

72

Figure 31. Infarct size across different treatment groups in the 8K-NBD study.

A cross-section from each heart was taken and apoptosis assessed by TUNEL staining.

The nuclei were stained with Dapi, and the number of TUNEL positive nuclei expressed as a

percentage of total nuclei in each field. The whole heart cross-section was taken into

consideration by taking 5-6 contiguous, non-overlapping sections from each heart. The figure

below demonstrates that the highest number of apoptotic nuclei occurred in 8K-NBD treated

hearts, followed by 8K-mutNBD hearts with least number seen in the PBS treated heart sections.

0.0

10.0

20.0

30.0

40.0

50.0

60.0

70.0

8K-NBDn=6

8K-mutant NBDn=6

PBSn=6

Treatment Groups

Perc

ent I

nfar

ct S

ize

p=0.10

p=0.001

Maliha
Placed Image
Maliha
Placed Image
Maliha
Placed Image
Maliha
Placed Image
Page 88: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

73

Figure 32. Representative confocal micrographs of TUNEL staining in each treatment group in the

8K-NBD study.

The figure below (Figure 33) illustrates the TUNEL staining data in a quantitative

manner.

8K-NBD 8K-mutNBD

PBS

Page 89: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

74

Figure 33. Quantification of TUNEL positive data in each treatment group in 8K-NBD study.

From these set of experiments we conclude that although 8K-NBD is able to inhibit NF-

κB activation in response to LPS stimulation, this inhibition in a mouse infarct model appears to

be detrimental and results in increased apoptosis occurring, as assessed by TUNEL staining, and

a larger infarct size. Although 8K-mutNBD had lesser number of apoptotic nuclei, the infarct

size was actually larger. This difference from the PBS treated group reached statistical

significance. It is possible that although 8K-mutNBD has much reduced NF-κB inhibitory

activity, it still has some level of inhibition which in an acute infarct model is detrimental. The

apoptosis being less with a larger infarct size is difficult to explain and possibly within the realm

-0.5

0

0.5

1

1.5

2

2.5

3

3.5

4

8K-NBD 8K-mutant NBD PBS

Per

cent

TU

NE

L P

ositi

ve N

ucle

i

Treatment Groups

Percent TUNEL Positive Nuclei in Whole Heart

Maliha
Placed Image
Page 90: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

75

of biological variability as the infarct size in 8K-NBD treated and 8K-mutNBD treated mice was

not statistically significant.

3.3 INFARCT STUDIES USING CTP-NBD

From the above section we conclude that although 8K-NBD is able to inhibit LPS-induced NF-

κB activity, such inhibition led to increased apoptosis and increase in infarct size. However the

nature of these apoptotic nuclei, white blood cells/infiltrating cells versus cardiomyocytes was

not clear. Data from this lab in a mouse model of inflammatory bowel disease in which treatment

with 8K-NBD peptide was efficacious [18]. From our phage display experiments, we also noted

that the transduction of heart tissue by 8K is quite inferior compared to CTP. We therefore

hypothesized that the superior transduction ability of CTP would lead to better delivery of the

NBD peptide to the heart tissue under ischemic stress and by inhibiting NF-κB activation above

its basal level would be cardioprotective and reduce infarct size.

Female, Balb/c mice underwent left artery ligation as detailed in Section 3.1 above. Mice

were treated either with PBS, CTP-NBD or 8K-NBD (both at a dose of 10mg/Kg body weight)

on day of surgery, delivered i.p., right after ischemia was confirmed. Peptides were suspended in

PBS. The surgeon was blinded to the treatment allocation and treatment was given in a rotational

fashion. Post-operative pain control was achieved with I/M Ketoprofen (5mg/Kg) on day 0, 1

and 2. Mice were euthanized on post-op day 7, heart dissected and processed for infarct size

determination as detailed in Section 3.1 above. The post-operative mortality in each group is

shown in the figure below (Figure 34).

Page 91: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

76

Figure 34. Post-operative mortality by treatment group in the CTP-NBD study.

The infarct size is shown in the figure below (Figure 35). As before, the infarct size was

slightly more in the 8K-NBD treated group compared to the PBS treated group. However the

CTP-NBD treated group showed a trend towards smaller infarct sizes.

Page 92: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

77

Figure 35. Infarct size by treatment groups in the CTP-NBD study.

Although this data was encouraging, the effect size was small and statistical analysis

using this data showed that a prohibitively large sample size in each group would be needed to

show significance. We decided to take an alternative approach. We hypothesized that the D-

isoform of CTP-NBD would be resistant to degradation, have a longer half-life and be able to

inhibit NF-κB activation above its basal level much more effectively. To test this hypothesis, the

D-isoform of CTP-NBD was synthesized in the University of Pittsburgh Peptide Synthesis

Facility, with a fraction biotinylated at the C-terminus for conjugation with Streptavidin-

Alexa488 (SA488) for tracking studies.

N=6

N=8 N=8

Page 93: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

78

Using the D-isoform of CTP-NBD (D-CTP-NBD) to reduce infarct studies raised a few

questions. Since the mechanism of transduction with CTP is not known, would switching the L-

isoform with the D-isoform affect the transduction abilities adversely? Also would the D-isoform

of NBD not be able to bind the IKK complex and therefore not inhibit NF-κB activation? To

answer these questions the following studies were undertaken prior to repeating the mouse

infarct studies with D-CTP-NBD.

Two hundred microliters of a 1mM solution of D-CTP-NBD, biotinylated form, was

incubated with 20ul of SA488 (2mg/ml stock solution) at room temperature (RT), light-protected

for 60 minutes. 1ml of PBS was also incubated with 20ul of SA488 under identical incubation

conditions. Female, Balb/c mice were anesthetized with i.p. Avertin and injected retro-orbitally

with the D-CTP-NBD-SA488 complex at a dose of 10mg/Kg or PBS+SA488. Mice were then

euthanized at 30 minutes (time to peak transduction with L-isoform CTP) or 120 minutes, hearts

taken, fixed in 2% paraformaldehyde at RT for 4 hrs, overnight 30% Sucrose at 4˚C, frozen in

liquid N2 chilled Isopentane and cryosectioned. PBS injected mice were euthanized at only 30

minutes. After cross-staining with DRAQ5, a nuclear stain, confocal microscopy was performed

to look for fluorescence. Confocal microscopy showed robust uptake of D-CTP-NBD-SA488

complex albeit at a slower rate than L-isoform CTP. There was beginning of transduction noted

at the edges with deeper tissue showing transduction at 120 minutes, as shown in figure below

(Figure 36).

Page 94: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

79

Figure 36. Transduction of the mouse heart by the D-CTP-NBD-SA488 complex.

To investigate if D-CTP-NBD retains its ability to inhibit NF-κB activation in response to

TNFα stimulation, the cell culture Luciferase studies were repeated. H9C2 cells were plated onto

96-well plates and double transfected with a plasmid carrying Luciferase under an NF-κB

promoter site as well as a Renilla expressing control plasmid. 24 hours post transfection, cells

were pre-treated with increasing concentrations of the D-CTP-NBD peptide and 30 minutes later

challenged with TNFα for 3 hours. After three hours, cells were washed, trypsinized, lysed and

Luciferase and Renilla activity quantified in a luminometer. All work was done in quadruplicate.

PBS+SA488 D-CTP-NBD-SA488 – 30mins

D-CTP-NBD – 120mins

Page 95: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

80

The figure below (Figure 37) shows that D-CTP-NBD is a less potent inhibitor of NF-κB

activation with any appreciable inhibition occurring only at the highest tested dose (400uM).

Figure 37. Inhibition of NF-κB activation by the D-isoform of CTP-NBD.

Although inhibition of NF-κB did occur, it was seen only at the highest tested dose.

Given that the transduction process was slowed down as seen in the in vivo imaging mouse study

(Figure 36), we hypothesized that perhaps 30 mins of incubation/lead time was not adequate with

D-CTP-NBD. However pre-incubating with 60 mins or 120 mins produced identical results with

~40% inhibition seen, only with the highest dose (400uM), as compared with the TNFα-

stimulated cells (results not shown). In spite of these findings, we choose to test the peptide in a

mouse infarct model, hypothesizing that although inhibition occurs only at the highest tested

dose, this dose might be achievable given the robust transduction seen at 120 mins as well as

0

20

40

60

80

100

120

140

NoRx Plasmid TNF-alpha D-CTP-NBD-50

D-CTP-NBD-100

D-CTP-NBD-200

D-CTP-NBD-400

Cor

rect

ed L

ucife

rase

/Ren

illa

Rat

io

Treatment Groups

Page 96: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

81

probable accumulation of D-CTP-NBD inside cardiomyocytes to a high enough concentration

given it’s resistance to intracellular degradation.

Mice underwent left artery ligation, as detailed above. Only one dose of D-CTP-NBD or

PBS was administered at time of surgery (5mg/Kg, i.p.), and mice were euthanized on post-op

day 7. The surgeon was blinded to the treatment allocation and post-op pain control was

achieved with s.q. Buprenorphine (0.1mg/Kg body weight). As shown in the figure below,

infarct size did not differ between the two groups.

Figure 38. Infarct sizes in the two treatment groups.

In addition, if infarct sizes of less than 10% were excluded, the mean infarct size in the

two groups was nearly identical leading us to conclude that treatment with D-CTP-NBD did not

have any significant effect on infarct size. Probably the most likely explanation for this lack of

0.010.020.030.040.050.060.070.0

D-CTP-NBD PBS

Infarct Size

Page 97: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

82

an effect, despite promising earlier data, is the lack of adequate inhibition of NF-κB activation by

the D-isoform of NBD. Although inhibition in cell culture studies, utilizing H9C2 cells, did

occur, it was apparent only at the highest dose tested (400uM). This level might not have been

achievable at the dose of 10mg/Kg body weight in vivo to produce a biological effect. In

addition, later studies of NF-κB inhibition [3, 105, and 106] have shown that infarct sizes are

similar in control and NF-κB inhibited mice, but remodeling is favorably affected in the latter

group. In our studies, mice were euthanized at post-op day 7, which would be too early to see

differences in post-infarct modeling or differences in left ventricular function.

In summary, these set of experiments demonstrated lack of a significant biologically

favorable effect of inhibiting NF-κB, utilizing 8K-NBD. In fact a trend towards worsening of

infarct size was apparent in multiple studies suggesting that the initial surge of NF-κB activation

that follows ischemic myocardium may have a protective role. Also 8K, as a protein transduction

domain, may be targeting cell types different than the actual myocytes and may be targeting

more of the inflammatory infiltrating cells. Inhibiting NF-κB in neutrophils, utilizing NBD, has

been shown to increase apoptosis of these cells in response to LPS stimulation [17]. Increasing

neutrophil apoptosis, at the earliest stage of the inflammatory response to myocardial ischemia,

may well be detrimental. Lack of a response of any kind to D-CTP-NBD in the subsequent

studies was likely not a failure of transduction with the D-CTP isoform but rather a lack of

adequate NF-κB inhibition by the D-isoform of NBD.

Page 98: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

83

4.0 SUMMARY, IMPLICATION OF OUR FINDINGS AND FUTURE DIRECTIONS

4.1 SUMMARY OF OUR FINDINGS

In this body of work we have utilized a combinatorial cell culture and in vivo phage display to

identify a peptide that targets rat cardiomyoblast cells (H9C2 cells) in vitro and mouse cardiac

tissue in vivo. This peptide, which we termed cardiac targeting peptide, or CTP, is able to deliver

a cargo peptide, NBD or nemo-binding domain to H9C2 cells in culture, and deliver

Streptavidin-Alexa488 complex to mouse cardiac tissue in vivo. The method of phage display

was first demonstrated by Smith [32], and in vivo phage display by Pasquallini and colleagues

[37]. Since the initial description of in vivo phage display, studies have been carried out to target

vasculature ligands of different organs demonstrating that the endothelial lining of various

organs is far from being homogenous. Such heterogeneity of vasculature ligands has been

demonstrated for different normal organs as well as diverse disease states like tumor vasculature

[37] and atherosclerotic plaques [126]. Most of these studies have focused on targeting vascular

ligands. In contrast, our work was meant to target more the cardiomyocytes in vivo, going

beyond the vasculature. We aimed to identify a peptide that would be able to traverse the

endothelial barrier of the heart, traverse the interstitium, bind and be internalized by

cardiomyocytes in vivo. Such a unique peptide would have to be teased out of a large library,

with high background of contaminating, non-specific phage, especially as the heart is an

Page 99: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

84

extremely vascular organ and would be rich in the circulating blood pool of phage. We optimized

our chances by pre-enriching the phage pool with a single cell culture cycle on H9C2 cells before

undertaking the in vivo cycles of phage display. The problem of contaminating phage in the

blood pool was minimized with prolonged circulation times to allow for clearance of phage from

the circulation and maximize the chances that phage recovered from the heart would be the

relevant, internalized one. Further, to maximize our chances of recovering phage, mice were pre-

treated with chloroquine to minimize degradation of internalized phage by preventing

acidification of the intracellular lysosomal compartment. Using these methodological changes,

we were able to identify CTP and show that it is not simply associated with vascular or

cardiomyocytes surface, but able to be internalized.

As a separate, though related, piece of work we went on to establish a mouse model of

myocardial infarction and test nemo-binding domain (NBD) peptide’s ability to reduce infarct

size in association with 8K, a known PTD from this lab. Later as the identity and transduction

ability of CTP was established, we utilized CTP-NBD to assess if this peptide was able to reduce

infarct size in vivo. As detailed in Chapter 3, though 8K-NBD was able to inhibit surge of NF-

κB activation in response to LPS, it actually increased infarct size in our model. Although CTP-

NBD was able to somewhat reduce infarct size in earlier studies, the effect size was modest and

would require large sample sizes to reach statistical significance. To prolong the action duration

of CTP-NBD, we utilized the D-isoform of all the amino acids constituting the peptide. Although

transduction ability of CTP was retained, it slowed down considerably and NBD was not able to

inhibit NF-κB activation, except at the highest concentration tested, and likely non-physiological

doses (400uM), in vitro. Such high concentrations were likely not achieved, in vivo, and may

explain the lack of effect noted with D-CTP-NBD.

Page 100: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

85

4.2 IMPLICATIONS AND FUTURE DIRECTIONS

A protein transduction domain able to transduce heart tissue specifically in vivo would open up

avenues for better imaging, as well as a delivery agent for potential therapeutics. We believe that

this is what we have achieved with identification of CTP. However this work has opened many

interesting questions. Foremost is the mode of transduction pathway that this peptide utilizes.

From the above discussion (Section 4.1), it is clear that this mode of delivery is likely to be

complex and has to overcome many barriers to actual transduction of the cardiomyocytes in vivo.

There has been immense interest in the mode of transduction utilized by PTDs in general with

greatest body of work gathered on TAT. This work has revealed that TAT, being a highly

cationic peptide, binds to heparan sulfate groups on cell surface followed by internalization,

likely by a macropinocytotic mechanism [127-128]. This is unlikely to be the mode of uptake by

CTP, as it is neither arginine-rich, nor is it cationic, with an isoelectric pH of 9.35 and

hydrophilicity index of -0.4. Indeed our lab has started performing preliminary experiments to

tease out the mechanism of transduction. In one set of preliminary experiments, H9C2 cells were

incubated with 200uM of CTP-SA488 complex at 37̊C or 4˚C for 30 mins. After equal number

of washes, cells were fixed, counter-stained with DRAQ5 and confocal microscopy performed.

The figure below (Figure 39) shows transduction occurring at 37˚C with minimal to no

transduction seen at 4̊C, implying that transduction is an active, energy -dependent process, and

not a simple diffusion of a peptide across the lipid cell membrane.

Page 101: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

86

Figure 39. Temperature dependency of CTP's transduction ability.

Moreover viability studies were performed in H9C2 cells after incubating with increasing

concentrations of CTP-NBD for 30 minutes at 37̊ C/5% CO2. After the incubation period, cells

were washed and viability assessed using a standard MTT assay. As shown in the graph below,

no significant change in viability was noted with CTP-NBD, even at the highest end of the tested

concentration. Therefore transduction is not likely simple uptake of CTP by compromised cells.

PBS

SA-488-37ºC

CTP+SA-488-37ºC

SA-488-4ºC

CTP+SA-488-4ºC

Page 102: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

87

Figure 40. H9C2 cell viability after incubating with increasing concentrations of CTP-NBD.

Interestingly a group of investigators have identified the identical CTP sequence, via

phage display, as binding preferentially to bone-like mineral and hydroxyapatite material [129-

130]. Although this work involved in vitro phage display against immobilized hydroxyapatite

discs, the fact that such a display led to the exact same sequence as CTP being identified

suggests that this peptide may be able to bind to cardiac interstitium. However for this to happen,

the heart endothelial barrier must be traversed first. Zhang and colleagues profiled heart

vasculature endothelium using in vivo phage display as well as bacterial two-hybridization

scheme, and identified a number of peptides able to bind anywhere from ~15 fold to 310 fold

greater avidity to heart endothelial cells [39]. None of these identified peptides bear any

Page 103: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

88

homology to the CTP sequence. Moreover, the high magnification confocal micrographs of

H9C2 cells (Figure 11) showing punctate intracellular, cytoplasmic fluorescence, along with 4˚C

transduction experiments reported above, suggest that transduction with CTP is an energy

dependent process involving binding of CTP to a ligand followed by endocytosis. The ligand

CTP is targeting remains to be identified. The next step would then be to confirm the

cytoplasmic substructure that CTP ends up in, using higher magnification confocal micrographs

of H9C2 cells along with endocytic markers. The exact ligand CTP is targeting can be identified

using a bacterial two-hybridization scheme similar to that reported by Zhang et. Al. [39]. Such

mechanistic insights would be critical to assess CTP’s ability to deliver cargoes, in addition to

the size limitations, to the heart. It is likely that the size and/or nature of the cargo could very

well diminish CTP’s transduction ability.

I see the potential clinical utility of CTP as not only a therapeutic delivery vehicle, which

of in itself would be of great benefit, but also as improving our diagnostic abilities. Recently

attention has focused on patient exposure to medically necessary ionizing radiation [131] which

can be considerable over time and have a cumulative effect. As was also pointed out in this study

[131], a major portion of the ionizing radiation exposure is from nuclear studies of which cardiac

nuclear studies constitute a significant percent. When injecting patients with radio-isotopes like

Thallium or Technetium, the majority of the radiation is taken up by the liver and gut regions.

Not only does that necessitate injecting a higher amount to target the heart, but also, not

infrequently, degrades the cardiac image quality. The inferior wall of the left ventricle is

particularly difficult to assess because of its proximity to the diaphragm as well as high

background gut activity just underneath the diaphragm. Labeling CTP with a radioisotope, like

technetium, has the potential to decrease total radiation exposure and actually increase image

Page 104: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

89

quality by directing all of the radiation to the heart. The proof of principal and ability to label

peptides with radio-isotopes and use for imaging has already been reported [132] and therefore is

feasible.

Another interesting potential application of CTP would be to express it on coat-protein of

adenoviral or adeno-associated virus to target viral particles to the heart. In this arena, we have

started work with AAV viral vectors in collaboration with Dr. Nakai’s lab. This lab has produced

a gutted out version of the AAV9 vector, termed the AAV1.9-3 vector, which lack heparan

binding sites. This viral vector has prolonged half-life in blood and limited transduction ability.

This virus can be retargeted by placing an RGD motif back in its coat protein. Preliminary

experiments assessing ability of AAV2, AAV1.9-3, AAV1.9-3RGD and AAV1.9-3-CTP (with

CTP sequence placed at position 587 of the viral coat protein) to transduce H9C2 cells appear

promising. The figure below demonstrates the ability to de-target and retarget AAV9 vector.

Addition of CTP to the de-targeted virus appears to be cautiously promising, though much work

remains to be done.

Page 105: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

90

AAV2 AAV1.3-9

AAV1.3-9-RGD AAV1.3-9-CTP

Figure 41. H9C2 cells transfected with different AAV viral vectors expressing EGFP.

Two major issues are the inability to package larger marker genes like LacZ into these

modified viruses, and the sharp drop in titers that occurs with modification of the coat protein

making large scale production difficult, if not impossible. These problems are in the process of

being addressed and will hopefully help in making AAV vectors more cardiac specific.

Page 106: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

91

5.0 METHODS

This chapter will outline in depth the step by step methods followed to do in vitro and in vivo

phage display. It will be followed by details of the cell transfection protocols as well as the

Luciferase read-out assay utilized to document TNFα mediated NF-κB activation as well as

inhibition of it by CTP-NBD fusion peptide. Later in the chapter, details will be provided of the

mouse infarct model, euthanasia protocol as well as determination of infarct size on H&E

histology slides.

5.1 PROTOCOL FOR PEPTIDE PHAGE DISPLAY

The protocol describes below the steps leading to a pre-screening cycle being performed in vitro

on rat cardiomyoblast cell line, or H9C2 cells, utilizing a commercially available (New England

Biolabs, cat#E8110S) M13, twelve amino acid peptide phage display library. After the first cycle

in vitro, the subsequent three cycles were performed in vivo, as detailed below.

The protocol detailed below, utilizing peptide phage display to identify internalizing

phage and the peptide sequence leading to internalization, is modified to target heart tissue in

vivo. The process is complicated by the heterogeneity and complexity of the tissue being

targeted, the sequestration of injected phage by organs of the reticuloendothelial system, largely

the liver and spleen, and a large pool of contaminating, non-specific circulating phage. In

Page 107: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

92

addition, the rigorous conditions that the phage displaying peptides are put through leads to loss

of infective phage over subsequent cycles. Here we provide details of the protocol used to

identify peptides transducing the heart with kidney being used as control tissue.

5.1.1 Materials

Cell Culture:

1. Dulbecco’s Modified Eagle’s Medium (DMEM; GIBCO/BRL)

2. Heat-inactivated fetal bovine serum (FBS; GIBCO/BRL)

3. Pen Strep stock solution (GIBCO; 15140)

4. Hepes buffer solution 1M (GIBCO; 15630)

5. Rat Cardiomyoblast Cell Line (H9C2 Cells) Media. Make the H9C2 cell media by adding

50ml of heat-inactivated FBS, 5ml of Hepes, and 5ml of Pen-Strep to 500ml of DMEM.

Filter the mixture and store at 4̊ C.

6. Solution of trypsin (0.25%) and ethylenediamine tetraacetic acid (EDTA-1mM; GIBCO)

Phage Display

1. M13 12-mer peptide phage display library containing:

a. Phage display peptide library, 100ul, ~1x1013 pfu/ml, in TBS with 50% glycerol

b. -96 gIII sequencing primer5´- HOCCC TCA TAG TTA GCG TAA CG –3´, 100

pmol, 1 pmol/μl

c. -28 gIII sequencing primer5´- HOGTA TGG GAT TTT GCT AAA CAA C –3´,

100 pmol, 1 pmol/μl

Page 108: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

93

d. E. coli ER2738. Host strain supplied as 50% glycerol culture; not competent.

Store at –70°C.

2. LB Medium: 10 g Bacto-Tryptone, 5 g yeast extract, 5 g NaCl in 1 liter of H2O.

Autoclave, store at 4̊ C.

3. IPTG/Xgal Stock Solution: Mix 1.25 g IPTG (isopropyl-β-D-thiogalactoside) and 1 g

Xgal (5-Bromo-4-chloro-3-indolyl-β-D-galactoside) in 25 ml DMF (dimethyl

formamide).Solution should be stored at –20°C.

4. LB/IPTG/Xgal Plates: 1 liter LB medium + 15 g/l agar. Autoclave, cool to < 70°C, add 1

ml IPTG/Xgal Stock per liter and pour.Store plates at 4°C in the dark.

5. Agar Top: 10gm Bacto-Tryptone, 5gm yeast extract, 5gm NaCl, 7gm Bacto-Agar in 1

liter of deiodized H2O. Autoclave, dispense into 50ml aliquots. Store at room temperature

(will be solid).

6. Tetracycline Stock Solution: 20mg/ml of tetracycline powder in a 1:1 Ethanol:Water.

Store at -20˚C, light-protected. Vortex before use.

7. LB+Tetracycline Media: To 1 liter of H2O add 10 g Bacto-Tryptone, 5 g yeast extract,

and 5 g NaCl. Autoclave, and allow to cool down to <70̊C or until lukewarm to touch.

Add 1ml of Tetracycline stock solution, mix and store at 4̊C, ligh t-protected, until ready

for use. Do not use if color turns from yellow to brown or black.

8. LB/Tetracycline Plates: Add 15gm of Agar to a liter of LB medium. Autoclave and allow

to cool to <70̊ C, add 1ml of tetracycline stock solution, pour into plates and allow to

solidify. Store plates at 4̊C, light -protected. Do not use plates if they turn brown or

black.

Page 109: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

94

9. TBS: 50 mM Tris-HCl (pH 7.5), 150 mM NaCl. Autoclave and store at room tempera-

ture.

10. PEG/NaCl: 20% (w/v) polyethylene glycol–8000, 2.5 M NaCl. Autoclave, mix well to

combine separated layers while still warm. Store at room temperature.

11. Iodide Buffer: 10 mM Tris-HCl (pH 8.0), 1 mM EDTA, 4 M sodium iodide (NaI).Store

at room temperature in the dark. Discard if color is evident.

Biopanning in mice

1. 8 to 12 week old albino, male or female mice (Balb/c or FVB; Jackson Labs)

2. 0.5ml Insulin syringes

3. Sterile phosphate buffered saline (PBS; GIBCO)

4. Avertin stock solution: Avertin (2, 2, 2-Tribromoethanol; Sigma-Aldrich) is mixed with

15. 5 ml tert-Amyl Alcohol (2-methyl-2-butanol; Fisher) for ~12 hours in a dark bottle at

room temperature. Allow the mixture to go completely into solution, sterile filter and

store at 4˚C, light protected. On the day of use, mix 200ul of this stock solution with

10ml of PBS, sterile filter and wrap in foil before use. This will be the 2% Avertin

working solution.

5. HBSS (Cellgro)

6. 10% Glucose. Dissolve 10gm of glucose in 100ml of deionized H2O, sterile filter in a

hood and store for later use

7. Heparin 1000U/ml (Lyphomed)

8. Collagenase II (50mg/ml; Worthington)

9. DNAse I (20mg/ml; Roche)

10. Dulbecco’s Modified Eagle’s Medium 50:50/F12 (Cellgro)

Page 110: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

95

11. Heat-inactivated fetal bovine serum (FBS; GIBCO/BRL)

12. Gentamycin 50mg/ml (1000x; GIBCO)

13. 75μM cell filters

2. Solution of trypsin (0.25%) and ethylenediamine tetraacetic acid (EDTA-1mM; GIBCO)

15. HBSS-G: To 90 ml of HBSS, add 10 ml of 10% glucose solution.

16. HBSS-G-Heparin: To 50ml of HBSS-G, add 500ul of Heparin (1000U/ml stock

solution). Make fresh on the day of use.

17. Digestion Enzyme: To 39ml of HBSS-G add 40ul of DNAse I and 1.2 ml of Collagenase II.

Make fresh on the day of use.

18. Plating medium: To 90 ml of DMEM 50:50/F12 add 10ml of the heat inactivated FBS and

100ul of Gentamycin stock solution.

19. 0.75% collagenase II: Add 75mg of collagenase II to 10ml of PBS. Make fresh on the day of

use.

20. DMEM 50:50/F12/25% FBS: add 7.5ml DMEM/F12 to 2.5ml FBS. Will need 10 cc for each

heart.

5.1.2 Methods

Peptide Phage Display

a. Passage the rat cardiomyoblast cell line, H9C2 cells, for a minimum of 3 passages (after

thawing from -180˚C). Cells should be passaged once they are ~70% confluent. Do not

allow to grow to complete confluency as they will begin to differentiate.

b. After the third passage, trypsinized cells with Trypsin/EDTA and plate in a 6-well plate

at a cell density of 2*105 cells/well. Twenty-four hours post-plating, aspirate and replace

Page 111: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

96

with pre-warmed media, and add 10ul of the M13 peptide phage library (~1011 phage)to

the media. Return cells to incubator for 6 hours.

c. After the incubation period, wash cell extensively (at least 6x) with pre-warmed PBS,

trypsinized cells, and centrifuge to collect cell pellet. Wash the cell pellet 1-2 times more

with media. Aspirate the supernatant media above the cell pellet but not completely. Cell

pellet may be stored at -80˚C for later lysis and phage titration.

d. Release internalized phage by subjecting the cell pellet to one freeze-thaw cycle by

bringing the cell pellet from -80˚C to either room temperature, or by placing in a 37̊ C

water-bath. Pellet out the cell debris and use the supernatant for phage titration as

detailed below.

Phage Titration

a. Inoculate 100ml LB+tetracycline media in a round-bottomed flask with 10ul of E. coli,

provided with the phage display library. Grow overnight at 37̊C with gentle shaking (225

rpm). This will be the starter culture.

b. Inoculate another 100ml of LB+tetracycline media with 1-2 drops of the starter culture.

Incubate at 37˚C for 1hr with gentle shaking.

c. Melt agarose top in a microwave until liquid. Vortex to make sure it is homogenous.

Dispense 3ml into 6 sterile culture tubes. Place the 6 culture tubes in a water-bath with

temperature set to 45̊ C and maintain at this temperature until ready for use.

d. To carry out infection, add 20 μl of cell extract (from Step 2.1d.) to 200ul of E. coli

culture (from Step 2.2b.), vortex quickly, and incubate at room temperature for 1–5

minutes. This will be the 1X100 dilution. Take 20ul of this and add to another 200ul of

Page 112: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

97

the starter culture (1X101) and so on until you have 1X106 dilutions. Use a new tip for

each serial dilution.

e. Transfer 200ul of each of these to the culture tubes containing 3ml of the melted agar top

at 45˚C, one infection at a time (from step 2.2d.), vortex briefly, and immediately pour

culture onto a pre-warmed LB+Tetracycline plate or IPTG/X-gal plate (labeled 1x100-----

----1x106). Pre-warm, for at least one hour, one LB/IPTG/Xgal plate per expected dilu-

tion at 37°C until ready for use. Gently tilt and rotate plate to spread top agar evenly.

f. Allow the plates to cool for 5 minutes, invert, and incubate overnight at 37°C. Place in

37˚C, light-protected (for either type of plates) for overnight growth.

g. Next day count either the clear plaques in the case of LB+Tetracycline plates, or blue

plaques in the case of Xgal plates. Some plates will have too numerous to count plaques

and some might have too few. Count the ones with 10-200 plaques, multiply by the

dilution factor and that will provide the number of phage per 20ul of the cell lysate. The

total number of phage can be calculated by multiplying this number by the total lysate

cell supernatant volume (in ul) divided by 20.

Total No of Phage = Number of plaques in a plate * Dilution factor of the plate *

total cell lysate volume/20

Phage Amplification

a. Inoculate 100ml LB+tetracycline media in a round-bottomed flask with 10ul of E. coli,

provided with the phage display library. Grow overnight at 37̊C with gentle shaking (225

rpm). This will be the starter culture.

Page 113: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

98

b. Inoculate another 100ml of LB+tetracycline media with 1-2 drops of the starter culture.

Incubate at 37˚C for 1hr with gentle shaking.

c. Take 32ml of this culture and add 1-1.5ml of the cell lysate (from 2d. above) to it and

grow for 4 hours at 37̊ C with gentle shaking (225 r pm).

d. Spin down at 10,000rpms at 4̊ C for 20minutes.

e. Discard the pellet. Take the 30ml of the supernatant and add 6ml of PEG/NaCl and keep

at 4˚C overnight.

f. Next day spin the tube at 12,000rpm for 20minutes at 4̊C.

g. Discard the supernatant and dissolve the pellet in 1cc of TBS (in the hood). Transfer to

eppendorf tube and add 200ul of PEG/NaCl (all in the hood). Place on ice for 1 hour.

h. Spin the tube in cold room at 14,000rpm for 5 minutes.

i. Discard the supernatant. Resuspend the pellet in 200ul of TBS. This is your amplified

phage. Titer this amplified phage using the protocol detailed in Methods-2.3. This will be

the amplified and tittered phage used for next cycle of phage display, which will after this

initial pre-screen, will be in vivo.

In vivo Peptide Phage Display

Weigh the mouse on a small animal scale and anesthetize using 12ul/gm of tissue weight of 2%

Avertin solution administered intraperitoneally. Mouse will be anesthetized in 2-5 minutes.

Inject 10ul of the peptide phage display library (~1011 pfu) intravenously, either retro-orbitally or

through a tail vein injection. The M13 phage, after an intravenous injection, has a half-life of

4.5hrs, in the circulation. Allow the mouse to recover and circulate the phage for the desired

number of half-lives (~3-6 half-lives).

Page 114: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

99

Isolating Cardiac Myocytes

a. After the desired time of circulation, re-anesthetize the mouse using 2% Avertin,

euthanize using either a CO2 chamber or cervical dislocation, and open the chest cavity.

Now place a nick in either the right atrium or right ventricle and using a small bore

needle, inject the left ventricle with 3-5ml of HBSS-G-Heparin, so as to flush out all the

red blood cells from the ventricular cavity and the coronary circulation.

b. Dissect out the heart, trim the atria and great vessels, and weigh the resulting trimmed

heart.

c. Place a petri dish on ice, add 2-4ml of HBSS-G-Heparin, place heart pieces in it and chop

as finely as possible.

d. Transfer all of this to a round bottom 5cc tube and discard the supernatant.

e. Add 2ml of Digestion Enzyme to the pieces, incubate (with rocking) at 37˚C for 5

minutes.

f. Collect the supernatant into DMEM 50:50/F12/25% FBS (10ml for each heart) media;

keep at 37˚C.

g. Repeat the steps e-f above until all the tissue pieces are digested.

h. Now place the accumulated tissue+DMEM+FBS through a 70µm filter.

i. Centrifuge the filtered cells for 4 minutes at 1000rpm.

b. Aspirate off the media, keep the pellet and resuspend the cells in 5-6ml of the plating

medium.

c. Now place in the cells and media in a 6” cell culture plate (to plate out the fibroblasts)

and place in an incubator at 37̊C for 2 hours (without disturbing).

Page 115: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

100

d. At the end of 2 hours, aspirate the top media, pellet the cells, resuspend in 1ml of PBS,

transfer to an eppendorf tube and place in -80˚C to freeze.

e. Put the cell pellet through one freeze-thaw cycle to lyse the cells and release the

internalized phage. Pellet the cell debris by centrifuging at maximum speed for 60 sec in

a table-top centrifuge. Use 20ul of this cell lysate/supernatant to titrate the released phage

as detailed in the phage titration section above. This phage population is the population of

interest that will be put through subsequent cycles of phage amplification (as detailed in

the phage amplification section above) and re-injected into a mouse for a second cycle of

peptide phage display. The number of phage recovered from the heart will be normalized

to the weight of the heart (in grams).

Isolating Kidney Cells

a. Inject phage library intravenously (reto-orbitally or via a tail vein injection) and allow to

circulate for the decided number of hours.

b. After the desired time of circulation, re-anesthetize the mouse using 2% Avertin,

euthanize using either a CO2 chamber or cervical dislocation, and open the chest cavity.

Now place a nick in either the right atrium or right ventricle and using a small bore

needle, inject the left ventricle with 3-5ml of HBSS-G-Heparin, so as to flush out all the

red blood cells from the ventricular cavity and the coronary circulation.

c. Dissect the kidney out and rinse in PBS.

d. In a cell culture hood, cut the kidney into small pieces with autoclaved/sterile razor blade.

e. Digest the minced tissue with 0.75% collagenase II at 37̊C for 30 -45minutes.

f. Pipette the digested tissue up and down.

Page 116: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

101

g. Filter the cells with 75µm filter.

h. Trypsinize the cells for 10minutes at room temperature.

i. Spin down the cells and wash with PBS twice.

j. Resuspend in 1ml of PBS.

k. Store in eppendorf tubes at -80˚C.

l. Put the cell pellet through one freeze-thaw cycle to lyse the cells and release the

internalized phage. Pellet the cell debris by centrifuging at maximum speed for 60 sec in

a table-top centrifuge. Use 20ul of this cell lysate/supernatant to titrate the released phage

as detailed in phage titration section above. This phage population is the control phage

and the released phage from heart, the target organ of interest, will be expressed as a ratio

of the number of phage recovered from kidney. The number of phage recovered from the

kidney will be normalized to the weight of the kidney (in grams).

m. After 3 cycles of in vivo peptide phage display, 10 plaques were picked from phage

recovered from the target organ, amplified and sequenced as detailed in Section “Phage

Sequencing” below. A consensus sequence emerged after 4 cycles of phage display,

which was termed “Cardiac Targeting Peptide” or CTP, and synthesized biotinylated,

labeled with 6-carboxyfluoroscein or as a fusion peptide with NBD or nemo-binding

domain peptide..

Phage Sequencing

From the plate with <100 plaques, pick 10-20 for amplification and subsequent

sequencing.

Page 117: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

102

a. Dilute an overnight culture of E. coli 1:100 in LB. Dispense 1 ml of diluted culture into

culture tubes, one for each clone to be characterized.

b. Use a sterile wooden stick or pipette tip to stab a plaque from a titering plate (important:

plates should be <1–3 days old, stored at 4°C and have <100 plaques) and transfer to a

tube containing the diluted culture. Pick well-separated plaques. This will ensure that

each plaque contains a single DNA sequence.

c. Incubate the tubes at 37°C with shaking for 4.5–5 hours (no longer).

d. Transfer the cultures to microcentrifuge tubes, and centrifuge at 14,000 rpm for 30

seconds. Transfer the supernatant to a fresh tube and re-spin. Using a pipette, transfer the

upper 80% of the supernatant to a fresh tube. This is the amplified phage stock and can be

stored at 4°C for several weeks with little loss of titer. For long-term storage (up to

several years), dilute 1:1 with sterile glycerol and store at –20°C.

e. Transfer 500 μl of the amplified phage stock to a fresh microfuge tube. Add 200 μl of

20% PEG/2.5 M NaCl. Invert several times to mix, and let stand for 10–20 minutes at

room temperature.

f. Microfuge at 14,000 rpm for 10 minutes at 4°C and discard the supernatant. Phage pellet

may not be visible. Re-spin briefly. Carefully pipet away and discard any remaining

supernatant.

g. Suspend the pellet thoroughly in 100 μl of Iodide Buffer by vigorously tapping the tube.

Add 250 μl of ethanol and incubate 10–20 minutes at room temperature. Short incubation

at room temperature will preferentially precipitate single-stranded phage DNA, leaving

most phage protein in solution.

Page 118: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

103

h. Spin in a microfuge at 14,000 rpm for 10 minutes at 4°C, and discard the supernatant.

Wash the pellet with 0.5 ml of 70% ethanol (stored at –20°C), re-spin, discard the

supernatant, and briefly dry the pellet under vacuum or at room air.

i. Resuspend the pellet in 30 μl of TE buffer. The template can be suspended in H2O

instead of TE if desired, but this is not recommended for long-term storage. In TE buffer,

the phage DNA should be stable indefinitely at –20°C. Sequence this DNA using the

sequencing primers provided with the peptide phage display library.

5.2 IN VITRO TRANSDUCTION EXPERIMENTS

5.2.1 Cell transduction experiments with CTP-6CF

In vitro cell transduction experiments were carried out using rat cardiomyoblasts (H9C2 cells),

mouse fibroblast cell line (NIH/3T3), mouse fibrosarcoma cell line (MCA205), human epithelial

cervical cancer cell line (HeLa cells) and human tubular kidney cell line (HK-2). Cells were

passaged at least three times and either grown to confluence or 70% confluence (in the case of

H9C2 cells) in the appropriate cell culture media. After at least 3 or more passages, cells were

trypsinized, counted on a hematocytometer and plated onto collagen-coated cover-slips at a cell

density of 2*105 cells/well in a 6-well tissue culture treated plate. Cells were left in the incubator,

37˚C/5% CO2 overnight. Next day media was aspirated, cells washed with pre-warmed 1ml of

Optimem and left in 1ml of Optimem. To the cells CTP-6CF was added at a final concentration

of 50, 100 or 200uM. A negative control group of incubating only with PBS was utilized. The

stock solution of the peptide was made in PBS at a 1mM concentration. After addition of the

Page 119: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

104

peptide, cells were returned to the incubator for 30 minutes. After the end of the incubation

period, media was aspirated, cells washed with pre-warmed PBS 3 times. After the washes, 1ml

of pre-warmed 2% paraformaldehyde was added and 1ul of DRAQ5 solution, a nuclear stain,

was added to the paraformaldehyde. This was carried out at RT for 30 minutes, light-protected.

After 30 minutes, paraformaldehyde was aspirated, cells washed with PBS once and cover-slips

mounted onto glass-slides using permount. The slides were kept at 4̊C, light -protected until next

day when confocal microscopy was performed.

For confocal microscopy, laser intensities/gains were set using negative control (PBS

injected) cells to minimize background fluorescence. Once the laser intensity for FITC was set, it

was kept constant across all subsequent imaging. Also serial scanning was performed to prevent

“bleed-through” from one laser wavelength to another.

As paraformaldehyde itself has auto-fluorescence, and boiling it can cause

polymerization of the individual molecules forming large complexes that can precipitate at lower

temperatures and cause fluorescence artifact, the protocol given below for making

paraformaldehyde was followed. The 8% stock solution was stored at 4̊C, light -protected,

wrapped in foil, and diluted 4:1 in PBS down to 2% on the day of use.

5.2.2 8% Paraformaldehyde stock solution

1. For each 100mls of Paraformaldehyde, add 8 grams of paraformaldehyde resin to 70 ml

of distilled water.

2. Heat to 70˚C (it will not dissolve).

3. Add 1M NaOH drop by drop until the solution clears; usually requires a couple of drops.

Page 120: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

105

4. Cool to room temperature.

5. Add 9mls of 1x PBS or 30 mls of 0.3M PBS

6. PH to 7.4

7. Filter and store at 4˚C, light-protected, wrapped in foil.

8. On the day of use, dilute necessary amount of 8% paraformaldehyde down to 2% with a

4:1 dilution with PBS.

5.3 METHODOLOGY FOR TRANSFECTION OF CELLS AND INHIBITION OF

NF-ΚB ACTIVATION

5.3.1 Materials

Materials specified are for 1 well of a 12-well plate. Scale up or down as necessary for

the size of the well and multiply by the number of wells needed. For example, one well of a 96-

well plate will need 6.25ul of Optimem. If the experiment is to be done in quadruplicate, would

need 25ul for each group in the experiment.

50ul of Optimem/well

2ul of Lipofectamine/well

200ng of reporter plasmid DNA/well (plasmid expressing Luciferase under an NF-κB

promoter site)

200ng of Renilla (control plasmid)/well

Page 121: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

106

5.3.2 Methods

1. After 3-4 cell passages, split cells using Trypsin/EDTA, and plate onto a 96-well plate at

a cell density of 2*104/well. Return cells to the incubator.

2. Next day incubate relevant amount of Optimem and with Lipofectamine at RT for 5 mins

(for every 50ul of Optimem use 2ul of Lipofectamine)..

3. Simultaneously incubate Optimem with the reporter plasmid DNA and the Renilla

control plasmid, (ratio of 50ul of Optimem with 200ng of reporter plasmid DNA and

200ng of Renilla control plasmid), at RT for 5 mins.

4. Add 1 and 2 together in a 1:1 ratio and incubate at RT for 20 minutes.

5. While above incubation is going on, aspirate media from cells, wash 3x with pre-warmed

antibiotic-free media and leave cells in 1ml of the antibiotic-free media (1ml for a 12-

well plate; 125ul for a 96-well plate).

6. Add 12.5ul of the optimem+lipofectamine+DNA into each well of a 96-well plate (100ul

per well of a 12-well plate) and leave in incubator o/n.

7. Next day, aspirate media, wash cells with pre-warmed Optimem once, and leave in 100ul

of Optimem (for a 96-well plate). Add the peptides in the concentrations being tested and

return cells to incubator for 30 minutes (or varying time-points as the experiment’s

hypothesis dictates).

8. Add murine (for H9C2 cells) or human TNFα (HeLa cells) at a concentration of 10ng/ml

and return cells to incubator for 3 hours.

Page 122: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

107

9. At the end of 3 hours, aspirate media, wash cells with pre-warmed PBS once, lyse cells

with 100ul of cell lysis buffer for 5 minutes. Pipette up and down a few times to complete

the lysis and add lysate to eppendorf tubes. Spin the debris down in a table-top

centrifuge, at maximum speed (usually 14,000 rpm) for 5 minutes, and transfer 10uleach

into two glass tubes for Luciferase and Renilla reading. The Luciferase and Renilla

activity is read in a Luminometer using Luciferase and Renilla substrate.

5.4 MOUSE INFARCT MODEL

The following protocol for generating infarcts in mice was utilized and kept consistent from one

set of experiments to another and between groups. Female, Balb/c, 10-20 week old mice were

used. Female mice were chosen because of the ability to house 5/cage, as opposed to male mice

(4/cage) and more importantly because female mice are able to tolerate cardiac insults better with

lower post-operative mortality [125]. Mice were anesthetized with intraperitoneal (i.p.) injection

of 2.5% Avertin (a mixture of 15.5 ml tert-amyl alcohol to 25 grams of 2-2-2 Tribromoethanol -

stock solution diluted to 2.5% concentration with PBS) at a dose of 15ul/gm of body weight.

Adequate anesthetic level was assessed by lack of response to toe-pinch and lack of a gag reflex.

Mice were intubated, using direct visualization of the vocal chords, with a 22G cannula and

placed on a Harvard rodent ventilator apparatus. Tube placement was confirmed by symmetric

expansion of the thoracic cavity. Mice were ventilated with ~0.1cc/breath tidal volume at a rate

of 110 breaths/minute.

Following successful intubation/ventilation, the chest cavity was opened using a left

lateral ternotomy approach. Care was taken not to extend the surgical incision cephalad to the

Page 123: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

108

level of the sternal head, as a large venous confluence is present in that region and cutting

through them can lead to major, exsanguinating bleed. A self-retaining mouse retractor was

placed inside the incision and the teeth opened to reveal the anterior surface of the heart. The

pericardium was gently removed with blunt forceps. A suture, 6-0 prolene, blue, monofilament

(Owens & Minor: #8714H) was placed along the lateral wall of the left ventricle (LV), ~2mm

below the lowest dip of the left atrium and ligated with exactly 5 squarely-placed knots. Ischemia

was confirmed by the resulting pallor of the myocardium and development of wall motion

abnormality. The muscle layer and skin were closed in two layers with running sutures with 8-0

black, monofilament nylon suture (Owens& Minor: #2808G). The mice were extubated once

spontaneous breathing was confirmed, and recovered on a heating pad. Post-operative pain was

managed with subcutaneous injection of Ketoprofen, 5mg/Kg, on post-op day 0, 1 and 2 or with

subcutaneous Buprenorphine, at a dose of 0.1mg/Kg body weight given once on the day of

surgery and twice daily on post-op day 1 and 2. Mice also got one injection of subcutaneous

Ampicillin, 100mg/Kg, after surgery for prophylaxis against post-surgical infection. Right after

ligation and suture placement on the anterior surface of the heart to produce myocardial

infarction, and before closing the chest wall, mice received i.p. peptides in different doses. The

surgeon (myself) was always blinded to the treatment. Also surgeries were carried out in a

rotational fashion so that any variation due to surgical technique was distributed equally over the

treatment groups.

Mice were euthanized on post-operative day 7. On day of euthanasia, mice were

anesthetized, chest cavity opened, and 1cc of 1M KCl injected via the right ventricle (RV) in

order to arrest the heart in diastole to prevent contraction band necrosis. This was followed by

injecting, through the right ventricle, 5cc of Glyo-fixx (formalin-substitute), heart dissected out

Page 124: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

109

and placed in 15ml of Glyo-fixx, until tissue embedded for sectioning. After paraffin embedding,

cross-sections of the heart were taken starting at the apex. The first section was taken once the

LV cavity appeared, with second and third sections taken 10 microns apart and cephalad of the

first section. The three sections were H&E stained, light microscopy photographs taken, and area

of infarct in each section mapped out using computer software Metamorph and expressed as a

percent of the whole heart. The infarct area from the three sections was averaged to give the final

infarct size for each mouse.

Page 125: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

110

BIBLIOGRAPHY

1. Wiviott, S.D., et al., Performance of the thrombolysis in myocardial infarction risk index in the National Registry of Myocardial Infarction-3 and -4: a simple index that predicts mortality in ST-segment elevation myocardial infarction. J Am Coll Cardiol, 2004. 44(4): p. 783-9.

2. Rogers, W.J., et al., Temporal trends in the treatment of over 1.5 million patients with myocardial infarction in the US from 1990 through 1999: the National Registry of Myocardial Infarction 1, 2 and 3. J Am Coll Cardiol, 2000. 36(7): p. 2056-63.

3. Kawano, S., et al., Blockade of NF-kappaB improves cardiac function and survival after myocardial infarction. Am J Physiol Heart Circ Physiol, 2006. 291(3): p. H1337-44.

4. Pachori, A.S., et al., Hypoxia-regulated therapeutic gene as a preemptive treatment strategy against ischemia/reperfusion tissue injury. Proc Natl Acad Sci U S A, 2004. 101(33): p. 12282-7.

5. Melo, L.G., et al., Molecular and cell-based therapies for protection, rescue, and repair of ischemic myocardium: reasons for cautious optimism. Circulation, 2004. 109(20): p. 2386-93.

6. Frankel, A.D. and C.O. Pabo, Cellular uptake of the tat protein from human immunodeficiency virus. Cell, 1988. 55(6): p. 1189-93.

7. Green, M. and P.M. Loewenstein, Autonomous functional domains of chemically synthesized human immunodeficiency virus tat trans-activator protein. Cell, 1988. 55(6): p. 1179-88.

8. Joliot, A.H., et al., alpha-2,8-Polysialic acid is the neuronal surface receptor of antennapedia homeobox peptide. New Biol, 1991. 3(11): p. 1121-34.

9. Fawell, S., et al., Tat-mediated delivery of heterologous proteins into cells. Proc Natl Acad Sci U S A, 1994. 91(2): p. 664-8.

10. Allinquant, B., et al., Downregulation of amyloid precursor protein inhibits neurite outgrowth in vitro. J Cell Biol, 1995. 128(5): p. 919-27.

11. Schwarze, S.R., et al., In vivo protein transduction: delivery of a biologically active protein into the mouse. Science, 1999. 285(5433): p. 1569-72.

12. Derossi, D., et al., The third helix of the Antennapedia homeodomain translocates through biological membranes. J Biol Chem, 1994. 269(14): p. 10444-50.

13. Pooga, M., et al., Cell penetration by transportan. FASEB J, 1998. 12(1): p. 67-77. 14. Morris, M.C., et al., A new peptide vector for efficient delivery of oligonucleotides into

mammalian cells. Nucleic Acids Res, 1997. 25(14): p. 2730-6. 15. Futaki, S., et al., Arginine-rich peptides. An abundant source of membrane-permeable

peptides having potential as carriers for intracellular protein delivery. J Biol Chem, 2001. 276(8): p. 5836-40.

Page 126: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

111

16. Mai, J.C., et al., Efficiency of protein transduction is cell type-dependent and is enhanced by dextran sulfate. J Biol Chem, 2002. 277(33): p. 30208-18.

17. Choi, M., et al., Inhibition of NF-kappaB by a TAT-NEMO-binding domain peptide accelerates constitutive apoptosis and abrogates LPS-delayed neutrophil apoptosis. Blood, 2003. 102(6): p. 2259-67.

18. Dave, S.H., et al., Amelioration of chronic murine colitis by peptide-mediated transduction of the IkappaB kinase inhibitor NEMO binding domain peptide. J Immunol, 2007. 179(11): p. 7852-9.

19. Ribeiro, M.M., et al., Heme oxygenase-1 fused to a TAT peptide transduces and protects pancreatic beta-cells. Biochem Biophys Res Commun, 2003. 305(4): p. 876-81.

20. Eguchi, A., et al., Protein transduction domain of HIV-1 Tat protein promotes efficient delivery of DNA into mammalian cells. J Biol Chem, 2001. 276(28): p. 26204-10.

21. Josephson, L., et al., High-efficiency intracellular magnetic labeling with novel superparamagnetic-Tat peptide conjugates. Bioconjug Chem, 1999. 10(2): p. 186-91.

22. Lewin, M., et al., Tat peptide-derivatized magnetic nanoparticles allow in vivo tracking and recovery of progenitor cells. Nat Biotechnol, 2000. 18(4): p. 410-4.

23. Rothbard, J.B., et al., Conjugation of arginine oligomers to cyclosporin A facilitates topical delivery and inhibition of inflammation. Nat Med, 2000. 6(11): p. 1253-7.

24. Torchilin, V.P., et al., TAT peptide on the surface of liposomes affords their efficient intracellular delivery even at low temperature and in the presence of metabolic inhibitors. Proc Natl Acad Sci U S A, 2001. 98(15): p. 8786-91.

25. Chiu, Y.L., et al., Visualizing a correlation between siRNA localization, cellular uptake, and RNAi in living cells. Chem Biol, 2004. 11(8): p. 1165-75.

26. Muratovska, A. and M.R. Eccles, Conjugate for efficient delivery of short interfering RNA (siRNA) into mammalian cells. FEBS Lett, 2004. 558(1-3): p. 63-8.

27. Moschos, S.A., et al., Lung delivery studies using siRNA conjugated to TAT(48-60) and penetratin reveal peptide induced reduction in gene expression and induction of innate immunity. Bioconjug Chem, 2007. 18(5): p. 1450-9.

28. Kumar, P., et al., Transvascular delivery of small interfering RNA to the central nervous system. Nature, 2007. 448(7149): p. 39-43.

29. Endoh, T., M. Sisido, and T. Ohtsuki, Cellular siRNA delivery mediated by a cell-permeant RNA-binding protein and photoinduced RNA interference. Bioconjug Chem, 2008. 19(5): p. 1017-24.

30. Lundberg, P., et al., Delivery of short interfering RNA using endosomolytic cell-penetrating peptides. FASEB J, 2007. 21(11): p. 2664-71.

31. Sergeeva, A., et al., Display technologies: application for the discovery of drug and gene delivery agents. Adv Drug Deliv Rev, 2006. 58(15): p. 1622-54.

32. Smith, G.P., Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. Science, 1985. 228(4705): p. 1315-7.

33. Scott, J.K. and G.P. Smith, Searching for peptide ligands with an epitope library. Science, 1990. 249(4967): p. 386-90.

34. Ackermann, H.W., Frequency of morphological phage descriptions in 1995. Arch Virol, 1996. 141(2): p. 209-18.

35. Ackermann, H.W., Frequency of morphological phage descriptions in the year 2000. Brief review. Arch Virol, 2001. 146(5): p. 843-57.

Page 127: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

112

36. Ackermann, H.W., 5500 Phages examined in the electron microscope. Arch Virol, 2007. 152(2): p. 227-43.

37. Arap, W., R. Pasqualini, and E. Ruoslahti, Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model. Science, 1998. 279(5349): p. 377-80.

38. Kolonin, M.G., et al., Reversal of obesity by targeted ablation of adipose tissue. Nat Med, 2004. 10(6): p. 625-32.

39. Zhang, L., J.A. Hoffman, and E. Ruoslahti, Molecular profiling of heart endothelial cells. Circulation, 2005. 112(11): p. 1601-11.

40. Pasqualini, R., D.M. McDonald, and W. Arap, Vascular targeting and antigen presentation. Nat Immunol, 2001. 2(7): p. 567-8.

41. Arap, W., et al., Steps toward mapping the human vasculature by phage display. Nat Med, 2002. 8(2): p. 121-7.

42. Mi, Z., et al., Identification of a synovial fibroblast-specific protein transduction domain for delivery of apoptotic agents to hyperplastic synovium. Mol Ther, 2003. 8(2): p. 295-305.

43. Rehman, K.K., et al., Protection of islets by in situ peptide-mediated transduction of the Ikappa B kinase inhibitor Nemo-binding domain peptide. J Biol Chem, 2003. 278(11): p. 9862-8.

44. Sen, R. and D. Baltimore, Inducibility of kappa immunoglobulin enhancer-binding protein Nf-kappa B by a posttranslational mechanism. Cell, 1986. 47(6): p. 921-8.

45. Sen, R. and D. Baltimore, Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell, 1986. 46(5): p. 705-16.

46. Makris, C., et al., Female mice heterozygous for IKK gamma/NEMO deficiencies develop a dermatopathy similar to the human X-linked disorder incontinentia pigmenti. Mol Cell, 2000. 5(6): p. 969-79.

47. Rudolph, D., et al., Severe liver degeneration and lack of NF-kappaB activation in NEMO/IKKgamma-deficient mice. Genes Dev, 2000. 14(7): p. 854-62.

48. Ting, A.T., F.X. Pimentel-Muinos, and B. Seed, RIP mediates tumor necrosis factor receptor 1 activation of NF-kappaB but not Fas/APO-1-initiated apoptosis. EMBO J, 1996. 15(22): p. 6189-96.

49. Blonska, M., et al., Restoration of NF-kappaB activation by tumor necrosis factor alpha receptor complex-targeted MEKK3 in receptor-interacting protein-deficient cells. Mol Cell Biol, 2004. 24(24): p. 10757-65.

50. Vallabhapurapu, S. and M. Karin, Regulation and function of NF-kappaB transcription factors in the immune system. Annu Rev Immunol, 2009. 27: p. 693-733.

51. Malinin, N.L., et al., MAP3K-related kinase involved in NF-kappaB induction by TNF, CD95 and IL-1. Nature, 1997. 385(6616): p. 540-4.

52. May, M.J., et al., Selective inhibition of NF-kappaB activation by a peptide that blocks the interaction of NEMO with the IkappaB kinase complex. Science, 2000. 289(5484): p. 1550-4.

53. May, M.J., R.B. Marienfeld, and S. Ghosh, Characterization of the Ikappa B-kinase NEMO binding domain. J Biol Chem, 2002. 277(48): p. 45992-6000.

54. Dai, S., et al., The IkappaB kinase (IKK) inhibitor, NEMO-binding domain peptide, blocks osteoclastogenesis and bone erosion in inflammatory arthritis. J Biol Chem, 2004. 279(36): p. 37219-22.

Page 128: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

113

55. di Meglio, P., A. Ianaro, and S. Ghosh, Amelioration of acute inflammation by systemic administration of a cell-permeable peptide inhibitor of NF-kappaB activation. Arthritis Rheum, 2005. 52(3): p. 951-8.

56. Shibata, W., et al., Cutting edge: The IkappaB kinase (IKK) inhibitor, NEMO-binding domain peptide, blocks inflammatory injury in murine colitis. J Immunol, 2007. 179(5): p. 2681-5.

57. De Plaen, I.G., et al., Inhibition of nuclear factor-kappaB ameliorates bowel injury and prolongs survival in a neonatal rat model of necrotizing enterocolitis. Pediatr Res, 2007. 61(6): p. 716-21.

58. Long, Y.M., et al., Cell-permeable Tat-NBD peptide attenuates rat pancreatitis and acinus cell inflammation response. World J Gastroenterol, 2009. 15(5): p. 561-9.

59. Ianaro, A., et al., NEMO-binding domain peptide inhibits proliferation of human melanoma cells. Cancer Lett, 2009. 274(2): p. 331-6.

60. Tapia, M.A., et al., Inhibition of the canonical IKK/NF kappa B pathway sensitizes human cancer cells to doxorubicin. Cell Cycle, 2007. 6(18): p. 2284-92.

61. Tas, S.W., et al., Local treatment with the selective IkappaB kinase beta inhibitor NEMO-binding domain peptide ameliorates synovial inflammation. Arthritis Res Ther, 2006. 8(4): p. R86.

62. van den Tweel, E.R., et al., Selective inhibition of nuclear factor-kappaB activation after hypoxia/ischemia in neonatal rats is not neuroprotective. Pediatr Res, 2006. 59(2): p. 232-6.

63. Biswas, D.K., et al., NF-kappa B activation in human breast cancer specimens and its role in cell proliferation and apoptosis. Proc Natl Acad Sci U S A, 2004. 101(27): p. 10137-42.

64. Ashikawa, K., et al., Evidence that activation of nuclear factor-kappaB is essential for the cytotoxic effects of doxorubicin and its analogues. Biochem Pharmacol, 2004. 67(2): p. 353-64.

65. Thomas, R.P., et al., Selective targeting of the nuclear factor-kappaB pathway enhances tumor necrosis factor-related apoptosis-inducing ligand-mediated pancreatic cancer cell death. Surgery, 2002. 132(2): p. 127-34.

66. Craig, R., et al., The cytoprotective effects of the glycoprotein 130 receptor-coupled cytokine, cardiotrophin-1, require activation of NF-kappa B. J Biol Chem, 2001. 276(40): p. 37621-9.

67. Hall, G., et al., Inhibitor-kappaB kinase-beta regulates LPS-induced TNF-alpha production in cardiac myocytes through modulation of NF-kappaB p65 subunit phosphorylation. Am J Physiol Heart Circ Physiol, 2005. 289(5): p. H2103-11.

68. Wright, G., et al., Endotoxin stress-response in cardiomyocytes: NF-kappaB activation and tumor necrosis factor-alpha expression. Am J Physiol Heart Circ Physiol, 2002. 282(3): p. H872-9.

69. Haudek, S.B., D.D. Bryant, and B.P. Giroir, Differential regulation of myocardial NF kappa B following acute or chronic TNF-alpha exposure. J Mol Cell Cardiol, 2001. 33(6): p. 1263-71.

70. Norman, D.A., M.H. Yacoub, and P.J. Barton, Nuclear factor NF-kappa B in myocardium: developmental expression of subunits and activation by interleukin-1 beta in cardiac myocytes in vitro. Cardiovasc Res, 1998. 39(2): p. 434-41.

Page 129: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

114

71. Pelzer, T., et al., Estrogen effects in the myocardium: inhibition of NF-kappaB DNA binding by estrogen receptor-alpha and -beta. Biochem Biophys Res Commun, 2001. 286(5): p. 1153-7.

72. Xuan, Y.T., et al., Nuclear factor-kappaB plays an essential role in the late phase of ischemic preconditioning in conscious rabbits. Circ Res, 1999. 84(9): p. 1095-109.

73. N'Diaye, M., et al., TNFalpha- and NF-kappaB-dependent induction of the chemokine CCL1 in human macrophages exposed to the atherogenic lipoprotein(a). Life Sci, 2009. 84(13-14): p. 451-7.

74. Sanchez-Galan, E., et al., Leukotriene B4 enhances the activity of nuclear factor-kappaB pathway through BLT1 and BLT2 receptors in atherosclerosis. Cardiovasc Res, 2009. 81(1): p. 216-25.

75. van Keulen, J.K., et al., The Nuclear Factor-kappa B p50 subunit is involved in flow-induced outward arterial remodeling. Atherosclerosis, 2009. 202(2): p. 424-30.

76. Gareus, R., et al., Endothelial cell-specific NF-kappaB inhibition protects mice from atherosclerosis. Cell Metab, 2008. 8(5): p. 372-83.

77. Crosara-Alberto, D.P., R.Y. Inoue, and C.R. Costa, FAK signalling mediates NF-kappaB activation by mechanical stress in cardiac myocytes. Clin Chim Acta, 2009. 403(1-2): p. 81-6.

78. Liang, Y.J., et al., Mechanical stress enhances serotonin 2B receptor modulating brain natriuretic peptide through nuclear factor-kappaB in cardiomyocytes. Cardiovasc Res, 2006. 72(2): p. 303-12.

79. Li, T., et al., MyD88-dependent nuclear factor-kappaB activation is involved in fibrinogen-induced hypertrophic response of cardiomyocytes. J Hypertens, 2009. 27(5): p. 1084-93.

80. Freund, C., et al., Requirement of nuclear factor-kappaB in angiotensin II- and isoproterenol-induced cardiac hypertrophy in vivo. Circulation, 2005. 111(18): p. 2319-25.

81. Young, D., et al., Blockade of NF-kappaB using IkappaB alpha dominant-negative mice ameliorates cardiac hypertrophy in myotrophin-overexpressed transgenic mice. J Mol Biol, 2008. 381(3): p. 559-68.

82. Gupta, S., et al., Prevention of cardiac hypertrophy and heart failure by silencing of NF-kappaB. J Mol Biol, 2008. 375(3): p. 637-49.

83. Ha, T., et al., Attenuation of cardiac hypertrophy by inhibiting both mTOR and NFkappaB activation in vivo. Free Radic Biol Med, 2005. 39(12): p. 1570-80.

84. Gupta, S., D. Young, and S. Sen, Inhibition of NF-kappaB induces regression of cardiac hypertrophy, independent of blood pressure control, in spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol, 2005. 289(1): p. H20-9.

85. Tillmanns, J., et al., Caught in the act: in vivo molecular imaging of the transcription factor NF-kappaB after myocardial infarction. Biochem Biophys Res Commun, 2006. 342(3): p. 773-4.

86. Maulik, N., et al., An essential role of NFkappaB in tyrosine kinase signaling of p38 MAP kinase regulation of myocardial adaptation to ischemia. FEBS Lett, 1998. 429(3): p. 365-9.

87. Meldrum, D.R., Mechanisms of cardiac preconditioning: ten years after the discovery of ischemic preconditioning. J Surg Res, 1997. 73(1): p. 1-13.

Page 130: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

115

88. Morgan, E.N., et al., An essential role for NF-kappaB in the cardioadaptive response to ischemia. Ann Thorac Surg, 1999. 68(2): p. 377-82.

89. Frantz, S., et al., Tissue-specific effects of the nuclear factor kappaB subunit p50 on myocardial ischemia-reperfusion injury. Am J Pathol, 2007. 171(2): p. 507-12.

90. Li, H.L., et al., Targeted cardiac overexpression of A20 improves left ventricular performance and reduces compensatory hypertrophy after myocardial infarction. Circulation, 2007. 115(14): p. 1885-94.

91. Trescher, K., et al., Inflammation and postinfarct remodeling: overexpression of IkappaB prevents ventricular dilation via increasing TIMP levels. Cardiovasc Res, 2006. 69(3): p. 746-54.

92. Trescher, K., et al., Adenovirus-mediated overexpression of inhibitor kappa B-alpha attenuates postinfarct remodeling in the rat heart. Eur J Cardiothorac Surg, 2004. 26(5): p. 960-7.

93. Hua, F., et al., Blocking the MyD88-dependent pathway protects the myocardium from ischemia/reperfusion injury in rat hearts. Biochem Biophys Res Commun, 2005. 338(2): p. 1118-25.

94. Sakaguchi, T., et al., A novel strategy of decoy transfection against nuclear factor-kappaB in myocardial preservation. Ann Thorac Surg, 2001. 71(2): p. 624-9; discussion 629-30.

95. Morishita, R., et al., In vivo transfection of cis element "decoy" against nuclear factor-kappaB binding site prevents myocardial infarction. Nat Med, 1997. 3(8): p. 894-9.

96. Misra, A., et al., Nuclear factor-kappaB protects the adult cardiac myocyte against ischemia-induced apoptosis in a murine model of acute myocardial infarction. Circulation, 2003. 108(25): p. 3075-8.

97. Meili-Butz, S., et al., Dimethyl fumarate, a small molecule drug for psoriasis, inhibits Nuclear Factor-kappaB and reduces myocardial infarct size in rats. Eur J Pharmacol, 2008. 586(1-3): p. 251-8.

98. Moss, N.C., et al., IKKbeta inhibition attenuates myocardial injury and dysfunction following acute ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol, 2007. 293(4): p. H2248-53.

99. Onai, Y., et al., Inhibition of IkappaB phosphorylation in cardiomyocytes attenuates myocardial ischemia/reperfusion injury. Cardiovasc Res, 2004. 63(1): p. 51-9.

100. Pye, J., et al., Proteasome inhibition ablates activation of NF-kappa B in myocardial reperfusion and reduces reperfusion injury. Am J Physiol Heart Circ Physiol, 2003. 284(3): p. H919-26.

101. Altavilla, D., et al., IRFI 042, a novel dual vitamin E-like antioxidant, inhibits activation of nuclear factor-kappaB and reduces the inflammatory response in myocardial ischemia-reperfusion injury. Cardiovasc Res, 2000. 47(3): p. 515-28.

102. Yeh, C.H., et al., Inhibition of NFkappaB activation with curcumin attenuates plasma inflammatory cytokines surge and cardiomyocytic apoptosis following cardiac ischemia/reperfusion. J Surg Res, 2005. 125(1): p. 109-16.

103. Yeh, C.H., et al., Inhibition of NF-kappa B activation can attenuate ischemia/reperfusion-induced contractility impairment via decreasing cardiomyocytic proinflammatory gene up-regulation and matrix metalloproteinase expression. J Cardiovasc Pharmacol, 2005. 45(4): p. 301-9.

Page 131: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

116

104. Timmers, L., et al., Targeted deletion of nuclear factor kappaB p50 enhances cardiac remodeling and dysfunction following myocardial infarction. Circ Res, 2009. 104(5): p. 699-706.

105. Wakatsuki, S., et al., A novel IKK inhibitor suppresses heart failure and chronic remodeling after myocardial ischemia via MMP alteration. Expert Opin Ther Targets, 2008. 12(12): p. 1469-76.

106. Frantz, S., et al., Absence of NF-kappaB subunit p50 improves heart failure after myocardial infarction. FASEB J, 2006. 20(11): p. 1918-20.

107. Squadrito, F., et al., Gene transfer of IkappaBalpha limits infarct size in a mouse model of myocardial ischemia-reperfusion injury. Lab Invest, 2003. 83(8): p. 1097-104.

108. Kupatt, C., et al., Retroinfusion of NFkappaB decoy oligonucleotide extends cardioprotection achieved by CD18 inhibition in a preclinical study of myocardial ischemia and retroinfusion in pigs. Gene Ther, 2002. 9(8): p. 518-26.

109. Hescheler, J., et al., Morphological, biochemical, and electrophysiological characterization of a clonal cell (H9c2) line from rat heart. Circ Res, 1991. 69(6): p. 1476-86.

110. Stathopoulou, K., I. Beis, and C. Gaitanaki, MAPK signaling pathways are needed for survival of H9c2 cardiac myoblasts under extracellular alkalosis. Am J Physiol Heart Circ Physiol, 2008. 295(3): p. H1319-H1329.

111. Saeedi, R., et al., Metabolic actions of metformin in the heart can occur by AMPK-independent mechanisms. Am J Physiol Heart Circ Physiol, 2008. 294(6): p. H2497-506.

112. Saeedi, R., et al., AMP-activated protein kinase influences metabolic remodeling in H9c2 cells hypertrophied by arginine vasopressin. Am J Physiol Heart Circ Physiol, 2009. 296(6): p. H1822-32.

113. Venkatakrishnan, C.D., et al., HSP27 regulates p53 transcriptional activity in doxorubicin-treated fibroblasts and cardiac H9c2 cells: p21 upregulation and G2/M phase cell cycle arrest. Am J Physiol Heart Circ Physiol, 2008. 294(4): p. H1736-44.

114. Turakhia, S., et al., Doxorubicin-induced cardiotoxicity: direct correlation of cardiac fibroblast and H9c2 cell survival and aconitase activity with heat shock protein 27. Am J Physiol Heart Circ Physiol, 2007. 293(5): p. H3111-21.

115. Molenaar, T.J., et al., Uptake and processing of modified bacteriophage M13 in mice: implications for phage display. Virology, 2002. 293(1): p. 182-91.

116. Ivanenkov, V., F. Felici, and A.G. Menon, Uptake and intracellular fate of phage display vectors in mammalian cells. Biochim Biophys Acta, 1999. 1448(3): p. 450-62.

117. Lundberg, M. and M. Johansson, Positively charged DNA-binding proteins cause apparent cell membrane translocation. Biochem Biophys Res Commun, 2002. 291(2): p. 367-71.

118. Lundberg, M., S. Wikstrom, and M. Johansson, Cell surface adherence and endocytosis of protein transduction domains. Mol Ther, 2003. 8(1): p. 143-50.

119. Gump, J.M. and S.F. Dowdy, TAT transduction: the molecular mechanism and therapeutic prospects. Trends Mol Med, 2007. 13(10): p. 443-8.

120. Madge, L.A. and M.J. May, Inhibiting proinflammatory NF-kappaB signaling using cell-penetrating NEMO binding domain peptides. Methods Mol Biol, 2009. 512: p. 209-32.

121. Bergmann, M.W., et al., Effect of NF-kappa B Inhibition on TNF-alpha-induced apoptosis and downstream pathways in cardiomyocytes. J Mol Cell Cardiol, 2001. 33(6): p. 1223-32.

Page 132: TARGETING THE HEART USING IN VIVO PHAGE DISPLAY

117

122. del Valle, J., et al., A new method for determining blood-brain barrier integrity based on intracardiac perfusion of an Evans Blue-Hoechst cocktail. J Neurosci Methods, 2008. 174(1): p. 42-9.

123. Rakos, G., et al., Evans Blue fluorescence permits the rapid visualization of non-intact cells in the perilesional rim of cold-injured rat brain. Acta Neurobiol Exp (Wars), 2007. 67(2): p. 149-54.

124. Kin, H., et al., Neutrophil depletion reduces myocardial apoptosis and attenuates NFkappaB activation/TNFalpha release after ischemia and reperfusion. J Surg Res, 2006. 135(1): p. 170-8.

125. Cavasin, M.A., et al., Gender differences in cardiac function during early remodeling after acute myocardial infarction in mice. Life Sci, 2004. 75(18): p. 2181-92.

126. Kelly, K.A., et al., In vivo phage display selection yields atherosclerotic plaque targeted peptides for imaging. Mol Imaging Biol, 2006. 8(4): p. 201-7.

127. Wadia, J.S., R.V. Stan, and S.F. Dowdy, Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nat Med, 2004. 10(3): p. 310-5.

128. Kaplan, I.M., J.S. Wadia, and S.F. Dowdy, Cationic TAT peptide transduction domain enters cells by macropinocytosis. J Control Release, 2005. 102(1): p. 247-53.

129. Segvich, S., et al., Identification of peptides with targeted adhesion to bone-like mineral via phage display and computational modeling. Cells Tissues Organs, 2009. 189(1-4): p. 245-51.

130. Segvich, S.J., H.C. Smith, and D.H. Kohn, The adsorption of preferential binding peptides to apatite-based materials. Biomaterials, 2009. 30(7): p. 1287-98.

131. Fazel, R., et al., Exposure to low-dose ionizing radiation from medical imaging procedures. N Engl J Med, 2009. 361(9): p. 849-57.

132. van den Borne, S.W., et al., Molecular imaging of interstitial alterations in remodeling myocardium after myocardial infarction. J Am Coll Cardiol, 2008. 52(24): p. 2017-28.